Published online Nov 28, 2024. doi: 10.3748/wjg.v30.i44.4689
Revised: October 25, 2024
Accepted: November 4, 2024
Published online: November 28, 2024
Processing time: 54 Days and 15.5 Hours
Pancreatic cancer (PC), a highly lethal tumor with nearly identical incidence and mortality rates, has become the sixth leading cause of cancer-related deaths. Hypoxia is an important malignant factor in PC, as it regulates angiogenesis, metabolic reprogramming, tumor progression, and metastasis. Disrupting the hypoxic microenvironment can enhance the efficacy of antitumor therapy and improve the prognosis of patients with PC. With the advent of bioinformatics, hypoxia-related PC models have emerged in recent years. They provide a referen
Core Tip: Currently, hypoxia-related bioinformatic models for pancreatic cancer (PC) primarily evaluate their value for prognosis, tumor microenvironment, and antitumor drug screening. However, these studies did not identify a prognostic model with an optimal predictive performance for PC. Moreover, findings based on bioinformatic analyses may not be completely reliable; thus, more experimental evidence is required. With the integration of multiomics data, the emergence of deep learning, and the application of high-quality experimental programs, the limitations of these models can be overcome and further application in clinical practice may be recommended in the future.
- Citation: Li DM, Cao XY, Jiang J. Hypoxia-related bioinformatic signatures associated with prognosis and tumor microenvironment of pancreatic cancer: Current status, concerns, and future perspectives. World J Gastroenterol 2024; 30(44): 4689-4696
- URL: https://www.wjgnet.com/1007-9327/full/v30/i44/4689.htm
- DOI: https://dx.doi.org/10.3748/wjg.v30.i44.4689
Pancreatic cancer (PC) is a leading cause of cancer-related deaths worldwide. Based on the latest cancer statistics from 2022, there were 510566 new cases of PC and 467005 deaths worldwide[1]. Of these, there were approximately 118700 new cases and 106300 deaths in China in 2022[2]. PC has become the third leading cause of cancer-related deaths in the United States, with an estimated 62200 new cases and 48800 deaths reported each year[3]. Although the incidence varies substantially among countries, global statistics indicate that PC is on the rise, and some scholars predict that it will soon become the second leading cause of cancer-related deaths in Western countries[3]. As a highly lethal and notorious disease, only 15%-20% of patients with PC are candidates for surgical resection. Because of the late diagnosis, rapid progression, early metastasis, and limited treatment options, the 5-year survival rate of PC is approximately 10%, thus posing a major threat to human health[4].
Unlike most solid tumors, the tumor environment of PC is characterized by abundant extracellular matrix rich in stromal cells but lacks extensive angiogenesis, resulting in continuous and severe intratumoral hypoxia[5]. As an important malignant hallmark, hypoxia is involved in the regulation of tumor growth, apoptosis, metabolism, stemness, progression, metastasis, and chemoresistance in PC through the activation of various molecules and signaling pathways[6-8]. Targeting the hypoxic microenvironment of PC is a novel antitumor treatment strategy. It may prevent aggressive progression, enhance the efficacy of chemoradiotherapy, and improve the prognosis of patients with PC.
With a better understanding of the hypoxic microenvironment in PC, researchers have begun to establish hypoxia-related bioinformatic signatures to evaluate prognosis, tumor microenvironment changes, and antitumor drug sensitivity of patients with PC. These signatures provide a reference for predicting the survival time and immune infiltration of patients with PC and provide potential biomarkers for targeting the hypoxic microenvironment of PC, which has attracted significant attention. In this editorial, we review the current status of hypoxia-related prognostic signatures in PC. Furthermore, we discuss several issues associated with these prognostic signatures and provide potential solutions and directions for further studies.
With an increased understanding of the hypoxic microenvironment in the oncology community, more than ten hypoxia-related bioinformatic models have been established to predict prognosis and tumor microenvironmental changes in patients with PC[9-19]. As shown in Table 1, these studies used hypoxia-related transcriptomic expression profiles and the corresponding clinical data from the Cancer Genome Atlas (TCGA) PC cohort as a training set to construct prognostic models based on Cox regression analysis and least absolute shrinkage and selection operator (LASSO) regression analysis. Subsequently, the accuracy of these models was tested using PC datasets from the Gene Expression Omnibus (GEO) and/or International Cancer Genome Consortium (ICGC) databases. Several common evaluation measures, such as Kaplan-Meier curves, receiver operating characteristic curves, C-index, and calibration curves, as well as univariate and multivariate prognostic risk factor analyses, were used to describe the predictive performance and robustness of the models. Overall, these prognostic models showed low-to-moderate predictive capability for a specific range of patients with PC.
Ref. | Data sources | Modeling methods | Biomarkers | Prognostic performance |
Yang et al[9] | TCGA, GSE62452 | Cox, LASSO | CAPN2, CCNA2, PLAU | 1-, 3-, 5-year area under the curve (AUC) in the training set: 0.687, 0.749, and 0.796; 1-, 3-, 5-year AUC in the test set: 0.610, 0.849, and 0.765; Calibration curve: Moderate fit; Better predictor than other clinical variables |
Ren et al[10] | TCGA, ICGC | Cox, LASSO | LY6D, PCAT2, RP11-80B9.1, RP3-525N10.2, TRIM67, UCA1 | 1-, 2-, 3-year AUC in the training set: 0.727, 0.911, and 0.93; 1-, 2-, 3-year AUC in test set 1: 0.635, 0.696 and 0.694; 1-, 2-, 3-year AUC in test set 2: 0.68, 0.756 and 0.689; Better predictor than other clinical variables; Independent prognostic factor |
Huang et al[11] | TCGA, ICGC, ArrayExpress E-MTAB-6134 | Cox, RSF | LDHA, POM121C | 1-, 3-, 5-year AUC in the training set: 0.716, 0.676, and 0.696; 1-, 3-, 5-year AUC in test set 1: 0.582, 0.642 and 0.657; 1-, 3-, 5-year AUC in test set 2: 0.711, 0.623 and 0.606; Better predictor than other clinical variables; Independent prognostic factor |
Li et al[12] | TCGA, GSE62452, GSE78229 | Cox, LASSO | KIF23, KRT13, LRP3, LY6D, MMP3, SERPINB7, SEC31B | 1-, 3-, 5-year AUC: 0.763, 0.832 and 0.814; Better predictor than other clinical variables; Independent prognostic factor |
Ren et al[13] | TCGA, ICGC | Cox, LASSO | ARID5A, FAM19A2, ICOSLG, IGLV7-46, SPRN | 1-, 2-, 3-year AUC in the training set: 0.77, 0.793, and 0.781; 1-, 2-, 3-year AUC in the test set: 0.675, 0.678 and 0.57; Better predictor than other clinical variables; Independent prognostic factor |
Zhou et al[14] | TCGA, GSE102238, GSE62452, GSE85916 | Cox, LASSO | BHLHE40, ENO1, SDC4, TGM2 | Calibration curve: Moderate fit; Independent prognostic factor |
Sun et al[15] | TCGA | Cox, LASSO | CCAT2, CEP83-DT, CYTOR, DANCR, GAS5, LINC01029, LINC01133, LINC01963, LINC02287, LINC-PINT, LNCSRLR, SH3PXD2A-AS1, TSPOAP1-AS1, UCA1 | 1-, 3-, 5-year AUC in the training set: 0.804, 0.89, and 0.915; 1-, 3-, 5-year AUC in the test set: 0.694, 0.769, and 0.866; Calibration curve: Moderate fit; Independent prognostic factor |
Tian et al[16] | TCGA, GSE62452 | Cox, LASSO | ANKZF1, CITED2, ENO3, JMJD6, LDHA, NDST1, SIAH2, TES | 1-, 3-, 5-year AUC in the training set: 0.936, 0.836, and 0.840; 1-, 3-, 5-year AUC in the test set: 0.814, 0.784, and 0.714; Calibration curve: Moderate fit; Independent prognostic factor |
Zhang et al[17] | TCGA, ICGC, GSE57495 | Cox | ANXA2, LDHA, TES | 1-, 3-, 5-year AUC in the training set: 0.683, 0.654, and 0.776; 1-, 3-, 5-year AUC in test set 1: 0.670, 0.628 and 0.761; 1-, 3-, 5-year AUC in test set 2: 0.684, 0.612 and 0.647; Independent prognostic factor |
Chen et al[18] | TCGA, GSE28735, GSE62452, ICGC | Cox, LASSO | GDF11, IL18, NR0B1, PLAU, PPP3CA, S100A16, SEMA3C | 1-, 3-, 5-year AUC in the training set: 0.76, 0.80, and 0.82; 1-, 3-, 5-year AUC in test set 1: 0.60, 0.83 and 0.79; 1-, 3-, 5-year AUC in test set 2: 0.75, 0.67 and 0.56; Calibration curve: Moderate fit; Better predictor than other clinical variables; Independent prognostic factor |
Ding et al[19] | TCGA, GSE78229, GSE57495 | Cox | ENO3, LDHA, PGK1, PGM1 | 1-, 3-, 5-year AUC in the training set: 0.701, 0.758, and 0.884; 1-, 3-, 5-year AUC in the test set: 0.602, 0.669, and 0.725; Independent prognostic factor |
In addition to measuring the predictive performance of these models, researchers also examined the correlation between the risk models and the tumor immune microenvironment (Table 2). Most studies have revealed that patients with high risk scores exhibit lower infiltration of antitumor CD8+ T cells, indicating the depletion of activated T cells and immune escape of tumor cells in the hypoxic microenvironment. Moreover, two studies reported that there may be a higher infiltration of M0 macrophages in the high-risk group[14,17]. Therefore, inducing the differentiation of these macrophages into antitumor M1 macrophages, rather than protumor M2 macrophages, may be necessary for high-risk patients. Nevertheless, the infiltrating levels of M2 macrophages in patients with high risk scores remain controversial based on the results of various hypoxia-related models. Ding et al[19] found that high-risk patients exhibited higher M2 macrophage infiltration, whereas the opposite finding was reported by Chen et al[18]. This contradiction can be attributed to the different backgrounds of the models and the algorithms used for the immune microenvironment.
Ref. | TME changes in a high-risk group | Therapeutic significance | Experimental validation | Mechanism |
Yang et al[9] | CD8+ T cells, T cells, B cells, plasmacytoid dendritic cells, immature dendritic cells, and cytotoxic cells | Higher sensitivity to cisplatin and paclitaxel in the high-risk group | qPCR: MRNA expression | None |
Ren et al[10] | CD8+ T cells, CD4+ T cells, and endothelial cells; Macrophages and fibroblasts | Higher sensitivity to paclitaxel, erlotinib, and cisplatin in the high-risk group; No significant difference in TMB, PD1, PD-L1, CTLA4 and IPS score | None | None |
Huang et al[11] | None | KRAS mutations are more frequent in the hypoxic subtype | qPCR: MRNA expression | None |
Li et al[12] | T cell exclusion scores | Lower PD1 expression in the high-risk subgroup | None | None |
Ren et al[13] | CD8+ T cells, B cells, macrophage, eosinophil, and monocyte; Immune activation score | Higher sensitivity to paclitaxel and erlotinib in the high-risk group; No significant difference in TMB, MSI, and IPS score | None | None |
Zhou et al[14] | CD8+ T cells, activated CD4+ memory T cells, naïve B cells, and plasma cells; M0 macrophages; Scores in CD4+ T cell recruiting, Th17 cell recruiting, dendritic cell recruiting, macrophage recruiting, and killing of cancer cells | None | qPCR: MRNA expression; Wound-healing assay: Migration; Transwell assay: Invasion | ChIP assay: BHLHE40/TLR3 axis |
Sun et al[15] | CD8+ T cells and B cells | None | qPCR: MRNA expression; MTT assay: Proliferation; Transwell assay: Invasion | ChIP assay and luciferase reporter assay: HIF-1α/ TSPOAP1-AS1 axis |
Tian et al[16] | CD8+ T cells and regulatory T cell | Lower PD1 and CTLA4 expression in a high-risk group | qPCR: MRNA expression | None |
Zhang et al[17] | T cells; M0 macrophages | Higher LDHA methylation in pancreatic cancer tissues | None | None |
Chen et al[18] | CD8+ T cells, follicular helper T cells, memory B cells, monocytes, M1 macrophages, M2 macrophages, resting mast cells, and eosinophils; Immune score and stromal score | Higher TMB in the high-risk groups | None | None |
Ding et al[19] | CD8+ T cells, plasma cells, and naïve B cells; M2 macrophages, resting memory CD4+ T cells, and resting natural killer cells | None | None | None |
These studies evaluated the roles of risk models in antitumor drug treatment, particularly immunotherapy (Table 2). Several important immune checkpoints (e.g., programed cell death protein 1, programed death ligand 1, and cytotoxic T-lymphocyte-associated protein 4), immunotherapy-related indicators (e.g., tumor mutation burden and microsatellite instability), and immunological scores (e.g., immunophenoscore and tumor immune dysfunction and exclusion score) were adopted to evaluate the relationship between risk models and immunotherapy response. Unfortunately, no significant findings regarding anticancer immunotherapy efficacy were observed in these studies; however, several chemotherapeutic and targeted drugs, including cisplatin, paclitaxel, and erlotinib, exhibited higher sensitivity in high-risk patients with PC, which to some extent, provide a reference for therapeutic decision-making in future clinical practice.
First, in these hypoxia-related bioinformatic models, the datasets used to construct prognostic signatures were entirely dependent on the TCGA cohort. Because of differences in ethnic and genetic background, data from a single source may fail to meet the universality of the model. Second, batch effects between different datasets should be avoided when building and testing these models, as there may be differences in transcriptomic data formats and sequencing methods. Different sources of raw data without uniform standardization may lead to an unreliable conclusion. Third, clear inclusion and exclusion criteria for patients with PC should be established to ensure the reproducibility of these models. Most deaths during the perioperative period were attributed to severe postoperative complications, such as pancreatic fistula and intraperitoneal infection, rather than PC progression. Therefore, patients who died in the short term after pancreatic surgery should be carefully considered for inclusion in model construction. Otherwise, the predictive performance of prognostic models may remain unchanged, with poor prediction accuracy for 1-year overall survival.
The ultimate goal of a study should not be limited to constructing a prognostic model with certain predictive effects. Instead, it should focus on continuously optimizing the model or identifying the model with the best predictive performance in PC. Because existing hypoxia-related prognostic models of PC were mainly constructed by routine Cox and LASSO regression analyses, they may not prove that the prognostic signature based on available data is the optimal model. Various machine learning algorithms have become increasingly popular and should be adopted to develop hypoxia-related models. Moreover, previous studies did not compare the predictive performance of these models.
Although almost all of these models were validated using external datasets, including GEO and ICGC data, the findings based on bioinformatic analysis may not be completely reliable; thus, they need to be validated with experimental evidence. Unfortunately, relevant experimental validation is not always performed well. Only a few studies have validated the relative mRNA expression levels of potential biomarkers that comprise prognostic models in PC cell lines. There is a lack of additional molecular biological, cytological, immunological, and animal experiments to verify the roles of these biomarkers in PC tumor growth, progression, metabolism, and immune microenvironment. A clinical PC cohort study has not been conducted to determine whether the expression of these biomarkers in tumor tissues affects the prognosis of patients with PC. An animal or hypoxic cell model should be established to clarify whether modulating the expression of these biomarkers can reverse the hypoxic microenvironment and improve PC prognosis. Moreover, the analysis of sensitivity and therapeutic response to anticancer drugs, including chemotherapy, targeted therapy, and immunotherapy, remains in a theoretical stage, as in vitro cytotoxicity assays and in vivo animal validation have not been reported. Overall, preclinical studies on relevant biomarkers in these prognostic models have substantial progress to make before their incorporation into clinical practice.
In addition to clarifying the roles of these prognostic biomarkers in PC, it is necessary to examine the pathways associated with biomarker function. Although multiple gene enrichment analyses have been used to identify risk model pathways in some studies, limited experimental evidence supports these findings. To date, only a few studies have used chromatin immunoprecipitation assays to evaluate the molecular interactions of selected biomarkers[14,15]. However, the mechanisms underlying the regulation of tumor progression, immune cell infiltration, and sensitivity to antitumor drugs through hypoxic microenvironments remain unclear. Notably, we found that lactate dehydrogenase A, a key enzyme that catalyzes the conversion of lactate to pyruvate and regulates lactylation, was repeatedly used when constructing prognostic signatures. The hypoxic microenvironment of PC is closely associated with abnormal metabolic repro
Although many hypoxia-related PC models have been constructed, no relevant clinical applications have been reported yet. This may be mainly attributed to the facts that the predictive accuracy and robustness of these prognostic signatures remain unsatisfactory and the model with the optimal predictive performance is not known. Meanwhile, unlike routine laboratory or clinical examinations of patients with PC, transcriptome sequencing of hypoxia-related genes in tumor tissues from patients with PC is time-consuming, complex, and expensive. In addition, there is currently a lack of more convincing experimental evidence to consider these predictive models for application in clinical practice. Moreover, for patients with advanced PC who do not undergo pancreatic surgery, we cannot predict survival or antitumor drug sensi
Single-cell and spatial transcriptomic sequencing have become important innovative technologies in biological research in recent years[23]. They are widely used to study gene expression at the single-cell level and the cell distribution and expression patterns at the spatial level. With the advent of single-cell and spatial transcriptomic technologies and the continuous breakthroughs in complex data processing and analysis, the construction of tumor prognostic models using single-cell and spatial transcriptomic analyses has become a hotspot in the field of bioinformatics[24,25]. On the one hand, single-cell and spatial transcriptomic data can be used to screen candidate genes in hypoxia-related models of PC. On the other hand, single-cell and spatial transcriptomic analyses may help us understand the expression characteristics of hypoxia-related genes at the single-cell and spatial levels. These analyses can also shed light on the immune microenvironment, cellular communication networks, and pathways associated with hypoxic conditions. Therefore, we should focus on analyzing the available single-cell and spatial transcriptome data for PC to provide new insights for the construction of prognostic signatures and identify the underlying mechanism associated with the hypoxic tumor microenvironment.
Hypoxia-related prognostic models based on traditional omics are analogous to a blind man being instructed to touch an elephant. Hence, accurately describing the prognostic landscape of the entire PC population is challenging. With breakthroughs in high-throughput sequencing technology, high-resolution mass spectrometry technology, and multiomics integration methods, systematic biomedical research has achieved a remarkable milestone[26]. Multiomics analysis can integrate genomics, epigenomics, transcriptomics, proteomics, metabolomics, pathomics, and radiomics in an unbiased manner to systematically analyze the underlying mechanisms of the hypoxic microenvironment and provide a novel paradigm for the establishment of hypoxia-related prognostic signatures. Multiomics integrated analysis and big-data health platforms will be used to construct predictive models for PC in the future. The combination of hypoxia-related multiomics data, clinical information, and radiological data of patients with PC is expected to achieve more accurate survival prediction and individualized treatment strategies, thereby effectively improving the prognosis and treatment of patients with PC.
Machine learning technology is a popular and cutting-edge technology in the field of artificial intelligence. It has powerful prediction, classification, and clustering capabilities to complete research tasks using various learning algorithms[27]. Deep learning is an emerging branch of machine learning that can simulate the learning process of the human brain by building multilayer neural network models, such as deep neural networks, convolutional neural networks, recurrent neural networks, and long short-term memory networks, to complete various complex tasks and achieve better prediction performance[28]. Recently, a study described the use of > 70 combinatorial machine learning algorithms to identify differentially expressed genes and construct a prognostic signature associated with programmed cell death in lung adenocarcinoma[29]. Similarly, another group integrated multiomics data and machine learning algorithms to identify three prognostic subtypes for invasive urothelial carcinoma and a consensus machine learning-driven signature with significant implications in prognosis and immunotherapy[30]. These signature-building strategies based on machine learning can provide a reference and experience for constructing hypoxia-related deep learning models in PC. With the continuous progress in computer science and technology, deep learning for the construction of prognostic models will develop rapidly. Using multiomics data and clinical information, various deep learning algorithms and their combinations can be applied to establish and continuously optimize hypoxia-related prognostic signatures until they achieve optimal predictive performance.
A convincing bioinformatic model requires external datasets to verify accuracy and robustness as well as experimental evidence for validation. Nevertheless, the current hypoxia-related models are not supported by biological experiments; thus, their reliability remains to be determined. We recommend the adoption of high-quality biological experiments in follow-up studies to examine the biological effects and mechanisms of hypoxia-related prognostic models in PC. For example, in addition to using quantitative real-time PCR and western blotting (WB) analyses to examine the expression of RNA and protein, fluorescence in situ hybridization, immunohistochemistry (IHC), and immunofluorescence (IF) are also good alternatives when validating the expression characteristics of hypoxia-related genes in PC. In addition, it is essential to establish a hypoxic PC cell model to examine the effect and mechanism of hypoxia-related genes on PC cell function. A co-culture system of PC cells and immune cells, such as CD8+ T cells and macrophages, should also be established under hypoxic conditions in future studies to examine how hypoxia-related genes influence immune cell function, such as cytokine production, cytotoxic activity, immune cell differentiation, and immune cell survival. Moreover, transplanted PC mouse tumor models should be established to study the role of hypoxia-related gene expression in the tumor immune microenvironment through molecular biological and immunological experiments, such as flow cytometry, IHC, IF, WB, and enzyme-linked immunosorbent assay. Common anticancer drugs and immune checkpoint inhibitors may also be administered to these in vivo models to determine the effect of hypoxia-related genes on the efficacy of chemotherapy, targeted therapy, and immunotherapy. Furthermore, the clinical cohorts of patients with PC should be established to verify the relationship between hypoxia-related gene expression in tumor tissues and prognosis and pathological progression.
Hypoxia-related bioinformatic models in PC are primarily evaluated in the context of prognosis, tumor immune microenvironment, and antitumor drug efficacy; however, these studies have not successfully identified a prognostic model with optimal predictive performance. Moreover, findings based on bioinformatic computational analyses are often not reliable, indicating that more experimental evidence is needed to identify the underlying mechanisms. With the integration of multiomics data, the application of deep learning algorithms, and the implementation of high-quality experimental programs, the limitations of these hypoxia-related prognostic models can be overcome, and they may be applied to clinical practice in the future.
1. | Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 72] [Cited by in F6Publishing: 1262] [Article Influence: 1262.0] [Reference Citation Analysis (0)] |
2. | Han B, Zheng R, Zeng H, Wang S, Sun K, Chen R, Li L, Wei W, He J. Cancer incidence and mortality in China, 2022. J Natl Cancer Cent. 2024;4:47-53. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 61] [Cited by in F6Publishing: 121] [Article Influence: 121.0] [Reference Citation Analysis (0)] |
3. | Stoffel EM, Brand RE, Goggins M. Pancreatic Cancer: Changing Epidemiology and New Approaches to Risk Assessment, Early Detection, and Prevention. Gastroenterology. 2023;164:752-765. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 48] [Reference Citation Analysis (0)] |
4. | Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet. 2020;395:2008-2020. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 869] [Cited by in F6Publishing: 1414] [Article Influence: 353.5] [Reference Citation Analysis (0)] |
5. | Tao J, Yang G, Zhou W, Qiu J, Chen G, Luo W, Zhao F, You L, Zheng L, Zhang T, Zhao Y. Targeting hypoxic tumor microenvironment in pancreatic cancer. J Hematol Oncol. 2021;14:14. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 68] [Cited by in F6Publishing: 212] [Article Influence: 70.7] [Reference Citation Analysis (0)] |
6. | Hao X, Ren Y, Feng M, Wang Q, Wang Y. Metabolic reprogramming due to hypoxia in pancreatic cancer: Implications for tumor formation, immunity, and more. Biomed Pharmacother. 2021;141:111798. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 9] [Cited by in F6Publishing: 33] [Article Influence: 11.0] [Reference Citation Analysis (0)] |
7. | Tan Z, Xu J, Zhang B, Shi S, Yu X, Liang C. Hypoxia: a barricade to conquer the pancreatic cancer. Cell Mol Life Sci. 2020;77:3077-3083. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 45] [Cited by in F6Publishing: 43] [Article Influence: 10.8] [Reference Citation Analysis (0)] |
8. | Yamasaki A, Yanai K, Onishi H. Hypoxia and pancreatic ductal adenocarcinoma. Cancer Lett. 2020;484:9-15. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 16] [Cited by in F6Publishing: 17] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
9. | Yang F, Jiang N, Li XY, Qi XS, Tian ZB, Guo YJ. Construction and validation of a pancreatic cancer prognostic model based on genes related to the hypoxic tumor microenvironment. World J Gastroenterol. 2024;30:4057-4070. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
10. | Ren M, Zhang J, Zong R, Sun H. A Novel Pancreatic Cancer Hypoxia Status Related Gene Signature for Prognosis and Therapeutic Responses. Mol Biotechnol. 2024;66:1684-1703. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 3] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
11. | Huang Y, Zhu Q, Sun Y, Zhang W, Zou J. Alterations in genes involved in glycolysis and hypoxia affect the prognosis of pancreatic cancer. Heliyon. 2024;10:e34104. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
12. | Li X, Yang X, Xue W, Yang R, He Z, Ai L, Liu H. Identification of gene signatures related to hypoxia and angiogenesis in pancreatic cancer to aid immunotherapy and prognosis. Front Oncol. 2023;13:1119763. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
13. | Ren M, Feng L, Zong R, Sun H. Novel prognostic gene signature for pancreatic ductal adenocarcinoma based on hypoxia. World J Surg Oncol. 2023;21:257. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
14. | Zhou L, Zhang W, Ni H, Liu J, Sun H, Liang Z, Wang R, Xue X, Chen K, Li W. A bioinformatics analysis and an experimental validation of the hypoxia-related prognostic model. J Gastrointest Oncol. 2023;14:1504-1524. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
15. | Sun J, Zhu S. Identifying the role of hypoxia-related lncRNAs in pancreatic cancer. Genomics. 2023;115:110665. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 2] [Reference Citation Analysis (0)] |
16. | Tian X, Zheng J, Mou W, Lu G, Chen S, Du J, Zheng Y, Chen S, Shen B, Li J, Wang N. Development and validation of a hypoxia-stemness-based prognostic signature in pancreatic adenocarcinoma. Front Pharmacol. 2022;13:939542. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 1] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
17. | Zhang JJ, Shao C, Yin YX, Sun Q, Li YN, Zha YW, Li MY, Hu BL. Hypoxia-Related Signature Is a Prognostic Biomarker of Pancreatic Cancer. Dis Markers. 2022;2022:6449997. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Reference Citation Analysis (0)] |
18. | Chen D, Huang H, Zang L, Gao W, Zhu H, Yu X. Development and Verification of the Hypoxia- and Immune-Associated Prognostic Signature for Pancreatic Ductal Adenocarcinoma. Front Immunol. 2021;12:728062. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 7] [Cited by in F6Publishing: 26] [Article Influence: 8.7] [Reference Citation Analysis (0)] |
19. | Ding J, He X, Cheng X, Cao G, Chen B, Chen S, Xiong M. A 4-gene-based hypoxia signature is associated with tumor immune microenvironment and predicts the prognosis of pancreatic cancer patients. World J Surg Oncol. 2021;19:123. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 10] [Cited by in F6Publishing: 11] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
20. | Nikanjam M, Kato S, Kurzrock R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol. 2022;15:131. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 240] [Reference Citation Analysis (0)] |
21. | Venturella M, Falsini A, Coppola F, Giuntini G, Carraro F, Zocco D, Chiesi A, Naldini A. CA-IX-Expressing Small Extracellular Vesicles (sEVs) Are Released by Melanoma Cells under Hypoxia and in the Blood of Advanced Melanoma Patients. Int J Mol Sci. 2023;24. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
22. | Wang W, Han Y, Jo HA, Lee J, Song YS. Non-coding RNAs shuttled via exosomes reshape the hypoxic tumor microenvironment. J Hematol Oncol. 2020;13:67. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 39] [Cited by in F6Publishing: 39] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
23. | Ahmed R, Zaman T, Chowdhury F, Mraiche F, Tariq M, Ahmad IS, Hasan A. Single-Cell RNA Sequencing with Spatial Transcriptomics of Cancer Tissues. Int J Mol Sci. 2022;23. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 24] [Cited by in F6Publishing: 31] [Article Influence: 15.5] [Reference Citation Analysis (0)] |
24. | Zhao S, Wang Q, Ni K, Zhang P, Liu Y, Xie J, Ji W, Cheng C, Zhou Q. Combining single-cell sequencing and spatial transcriptome sequencing to identify exosome-related features of glioblastoma and constructing a prognostic model to identify BARD1 as a potential therapeutic target for GBM patients. Front Immunol. 2023;14:1263329. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 11] [Reference Citation Analysis (0)] |
25. | Liu J, Li H, Wang L, Wang S, Tang Q. Spatial transcriptome and single-cell reveal the role of nucleotide metabolism in colorectal cancer progression and tumor microenvironment. J Transl Med. 2024;22:702. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
26. | He X, Liu X, Zuo F, Shi H, Jing J. Artificial intelligence-based multi-omics analysis fuels cancer precision medicine. Semin Cancer Biol. 2023;88:187-200. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 35] [Article Influence: 35.0] [Reference Citation Analysis (0)] |
27. | Swanson K, Wu E, Zhang A, Alizadeh AA, Zou J. From patterns to patients: Advances in clinical machine learning for cancer diagnosis, prognosis, and treatment. Cell. 2023;186:1772-1791. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 118] [Reference Citation Analysis (0)] |
28. | Tran KA, Kondrashova O, Bradley A, Williams ED, Pearson JV, Waddell N. Deep learning in cancer diagnosis, prognosis and treatment selection. Genome Med. 2021;13:152. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 275] [Cited by in F6Publishing: 270] [Article Influence: 90.0] [Reference Citation Analysis (0)] |
29. | Wang S, Wang R, Hu D, Zhang C, Cao P, Huang J. Machine learning reveals diverse cell death patterns in lung adenocarcinoma prognosis and therapy. NPJ Precis Oncol. 2024;8:49. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
30. | Chu G, Ji X, Wang Y, Niu H. Integrated multiomics analysis and machine learning refine molecular subtypes and prognosis for muscle-invasive urothelial cancer. Mol Ther Nucleic Acids. 2023;33:110-126. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 10] [Reference Citation Analysis (0)] |