1
|
Yan X, Gao X, Dong J, Wang F, Jiang X, Hu X, Zhang J, Wang N, Xu L, Liu Z, Hu S, Zhao H. Integration of Single-Cell and Bulk RNA-seq Data to Identify the Cancer-Associated Fibroblast Subtypes and Risk Model in Glioma. Biochem Genet 2025; 63:1275-1297. [PMID: 38536568 DOI: 10.1007/s10528-024-10751-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the stroma. Studies showed that CAFs were pivotally in glioma progression which have long been considered a promising therapeutic target. Therefore, the identification of prognostic CAF markers might facilitate the development of novel diagnostic and therapeutic approaches. A total of 1333 glioma samples were obtained from the TCGA and CGGA datasets. The EPIC, MCP-counter, and xCell algorithms were used to evaluate the relative proportion of CAFs in glioma. CAF markers were identified by the single-cell RNA-seq datasets (GSE141383) from the Tumor Immune Single-Cell Hub database. Unsupervised consensus clustering was used to divide the glioma patients into different distinct subgroups. The least absolute shrinkage and selection operator regression model was utilized to establish a CAF-related signature (CRS). Finally, the prognostic CAF markers were further validated in clinical specimens by RT‒qPCR. Combined single-cell RNA-seq analysis and differential expression analysis of samples with high and low proportions of CAFs revealed 23 prognostic CAF markers. By using unsupervised consensus clustering, glioma patients were divided into two distinct subtypes. Subsequently, based on 18 differentially expressed prognostic CAF markers between the two CAF subtypes, we developed and validated a new CRS model (including PCOLCE, TIMP1, and CLIC1). The nomogram and calibration curves indicated that the CRS was an accurate prognostic marker for glioma. In addition, patients in the high-CRS score group had higher immune infiltration and tumor mutation burden levels. Moreover, the CRS score had the potential to predict the response to immune checkpoint blockade (ICB) therapy and chemotherapy. Finally, the expression profiles of three CAF markers were verified by RT‒qPCR. In general, our study classified glioma patients into distinct subgroups based on CAF markers, which will facilitate the development of individualized therapy. We also provided insights into the role of the CRS in predicting the response to ICB and chemotherapy in glioma patients.
Collapse
Affiliation(s)
- Xiuwei Yan
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Gao
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Dong
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fang Wang
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyan Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xueyan Hu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
| | - Jiheng Zhang
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Wang
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Xu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
| | - Zhihui Liu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China.
| | - Shaoshan Hu
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Hongtao Zhao
- Cancer Center, Department of Neurosurgery, Hangzhou Medical College, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Liao J, Duan Y, Xu X, Liu Y, Zhan C, Xiao G. Circadian rhythm related genes signature in glioma for drug resistance prediction: a comprehensive analysis integrating transcriptomics and machine learning. Discov Oncol 2025; 16:119. [PMID: 39909964 PMCID: PMC11799505 DOI: 10.1007/s12672-025-01863-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/03/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Gliomas, 24% of all primary brain tumors, have diverse histology and poor survival rates, with about 70% recurring due to acquired or de novo resistance. Insomnia in patients is correlated strongly with circadian rhythm disruptions. The correlation between circadian rhythm disorders and drug resistance of some tumors has been proved. However, the precise mechanism underlying the relationship between glioma and circadian rhythm disorders has not been elucidated. METHODS Circadian rhythm-related genes (CRRGs) were identified using the least absolute shrinkage and selection operator (LASSO) regression, and stochastic gradient descent (SGD) was performed to form a circadian rhythm-related score (CRRS) model. The studies of immune cell infiltration, genetic variations, differential gene expression pattern, and single cell analysis were performed for exploring the mechanisms of chemotherapy resistance in glioma. The relationship between CRRGs and chemosensitivity was also confirmed by IC 50 (half maximal inhibitory concentration) analysis. RESULT Signatures of 16 CRRGs were screened out and identified. Based on the CRRS model, an optimal comprehensive nomogram was created, exhibiting a favorable potential for predicting drug resistance in samples. Immune infiltration, cell-cell communication, and single cell analysis all indicated that high CRRS group was closely related to innate immune cells. IC50 analysis showed that CRRG knockdown enhanced the chemosensitivity of glioma. CONCLUSION A significant correlation between CRRGs, drug resistance of glioma, and innate immune cells was found, which might hold a significant role in the drug resistance of glioma.
Collapse
Affiliation(s)
- Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxing Duan
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Xiangwang Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaxue Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chaohong Zhan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Körner M, Spohn M, Schüller U, Bockmayr M. Transcriptomics-based characterization of the immuno-stromal microenvironment in pediatric low-grade glioma. Oncoimmunology 2024; 13:2386789. [PMID: 39135890 PMCID: PMC11318680 DOI: 10.1080/2162402x.2024.2386789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Immunologic treatment options are uncommon in low-grade gliomas, although such therapies might be beneficial for inoperable and aggressive cases. Knowledge of the immune and stromal cells in low-grade gliomas is highly relevant for such approaches but still needs to be improved. Published gene-expression data from 400 low-grade gliomas and 193 high-grade gliomas were gathered to quantify 10 microenvironment cell populations with a deconvolution method designed explicitly for brain tumors. First, we investigated general differences in the microenvironment of low- and high-grade gliomas. Lower-grade and high-grade tumors cluster together, respectively, and show a general similarity within and distinct differences between these groups, the main difference being a higher infiltration of fibroblasts and T cells in high-grade gliomas. Among the analyzed entities, gangliogliomas and pleomorphic xanthoastrocytomas presented the highest overall immune cell infiltration. Further analyses of the low-grade gliomas presented three distinct microenvironmental signatures of immune cell infiltration, which can be divided into T-cell/dendritic/natural killer cell-, neutrophilic/B lineage/natural killer cell-, and monocytic/vascular/stromal-cell-dominated immune clusters. These clusters correlated with tumor location, age, and histological diagnosis but not with sex or progression-free survival. A survival analysis showed that the prognosis can be predicted from gene expression, clinical data, and a combination of both with a support vector machine and revealed the negative prognostic relevance of vascular markers. Overall, our work shows that low- and high-grade gliomas can be characterized and differentiated by their immune cell infiltration. Low-grade gliomas cluster into three distinct immunologic tumor microenvironments, which may be of further interest for upcoming immunotherapeutic research.
Collapse
Affiliation(s)
- Meik Körner
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
| | - Michael Spohn
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Bockmayr
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- bAIome - Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
4
|
Xu J, Zhang J, Chen W, Ni X. The tumor-associated fibrotic reactions in microenvironment aggravate glioma chemoresistance. Front Oncol 2024; 14:1388700. [PMID: 38863628 PMCID: PMC11165034 DOI: 10.3389/fonc.2024.1388700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
Malignant gliomas are one of the most common and lethal brain tumors with poor prognosis. Most patients with glioblastoma (GBM) die within 2 years of diagnosis, even after receiving standard treatments including surgery combined with concomitant radiotherapy and chemotherapy. Temozolomide (TMZ) is the first-line chemotherapeutic agent for gliomas, but the frequent acquisition of chemoresistance generally leads to its treatment failure. Thus, it's urgent to investigate the strategies for overcoming glioma chemoresistance. Currently, many studies have elucidated that cancer chemoresistance is not only associated with the high expression of drug-resistance genes in glioma cells but also can be induced by the alterations of the tumor microenvironment (TME). Numerous studies have explored the use of antifibrosis drugs to sensitize chemotherapy in solid tumors, and surprisingly, these preclinical and clinical attempts have exhibited promising efficacy in treating certain types of cancer. However, it remains unclear how tumor-associated fibrotic alterations in the glioma microenvironment (GME) mediate chemoresistance. Furthermore, the possible mechanisms behind this phenomenon are yet to be determined. In this review, we have summarized the molecular mechanisms by which tumor-associated fibrotic reactions drive glioma transformation from a chemosensitive to a chemoresistant state. Additionally, we have outlined antitumor drugs with antifibrosis functions, suggesting that antifibrosis strategies may be effective in overcoming glioma chemoresistance through TME normalization.
Collapse
Affiliation(s)
- Jiaqi Xu
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ji Zhang
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wubing Chen
- Department of Radiology, Wuxi Fifth People’s Hospital, Jiangnan University, Wuxi, China
| | - Xiangrong Ni
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Liu Z, Yang L, Xie Z, Yu H, Gu T, Shi D, Cai N, Zhuo S. Multi-cohort comprehensive analysis unveiling the clinical value and therapeutic effect of GNAL in glioma. Oncol Res 2024; 32:965-981. [PMID: 38686055 PMCID: PMC11055992 DOI: 10.32604/or.2024.045769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/21/2023] [Indexed: 05/02/2024] Open
Abstract
Clinical data indicates that glioma patients have poor treatment outcomes and clinical prognosis. The role of olfactory signaling pathway-related genes (OSPRGs) in glioma has not been fully elucidated. In this study, we aimed to investigate the role and relationship between OSPRGs and glioma. Univariate and multivariate Cox regression analyses were performed to assess the relationship between OSPRGs and the overall survival of glioma based on public cohorts, and the target gene (G Protein Subunit Alpha L, GNAL) was screened. The association of GNAL expression with clinicopathological characteristics, gene mutation landscape, tumor immune microenvironment (TIME), deoxyribonucleic acid (DNA) methylation, and naris-occlusion controlled genes (NOCGs) was performed. Immunohistochemistry was used to evaluate GNAL level in glioma. Further analysis was conducted to evaluate the drug sensitivity, immunotherapy response, and functional enrichment of GNAL. GNAL was an independent prognostic factor, and patients with low GNAL expression have a poor prognosis. Expression of GNAL was closely associated with clinicopathological characteristics, DNA methylation, and several immune-related pathways. Immune infiltration analysis indicated that GNAL levels were negatively correlated with immune scores. GNAL low-expression group showed efficacy with anti-PD-1 therapy. Ten compounds with significantly different half-maximal inhibitory concentration (IC50) values between the GNAL high and low-expression groups were identified. Furthermore, its expression was associated with several immune cells, immune-related genes, and NOCGs. The expression of GNAL is closely associated with clinicopathological characteristics, TIME, and the response to therapeutic interventions, highlighting its potential as a prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Liangwang Yang
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Zhengxing Xie
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Hui Yu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Tianyi Gu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Daoming Shi
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Ning Cai
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Shenghua Zhuo
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| |
Collapse
|
6
|
Manou D, Golfinopoulou MA, Alharbi SND, Alghamdi HA, Alzahrani FM, Theocharis AD. The Expression of Serglycin Is Required for Active Transforming Growth Factor β Receptor I Tumorigenic Signaling in Glioblastoma Cells and Paracrine Activation of Stromal Fibroblasts via CXCR-2. Biomolecules 2024; 14:461. [PMID: 38672477 PMCID: PMC11048235 DOI: 10.3390/biom14040461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Serglycin (SRGN) is a pro-tumorigenic proteoglycan expressed and secreted by various aggressive tumors including glioblastoma (GBM). In our study, we investigated the interplay and biological outcomes of SRGN with TGFβRI, CXCR-2 and inflammatory mediators in GBM cells and fibroblasts. SRGN overexpression is associated with poor survival in GBM patients. High SRGN levels also exhibit a positive correlation with increased levels of various inflammatory mediators including members of TGFβ signaling pathway, cytokines and receptors including CXCR-2 and proteolytic enzymes in GBM patients. SRGN-suppressed GBM cells show decreased expressions of TGFβRI associated with lower responsiveness to the manipulation of TGFβ/TGFβRI pathway and the regulation of pro-tumorigenic properties. Active TGFβRI signaling in control GBM cells promotes their proliferation, invasion, proteolytic and inflammatory potential. Fibroblasts cultured with culture media derived by control SRGN-expressing GBM cells exhibit increased proliferation, migration and overexpression of cytokines and proteolytic enzymes including CXCL-1, IL-8, IL-6, IL-1β, CCL-20, CCL-2, and MMP-9. Culture media derived by SRGN-suppressed GBM cells fail to induce the above properties to fibroblasts. Importantly, the activation of fibroblasts by GBM cells not only relies on the expression of SRGN in GBM cells but also on active CXCR-2 signaling both in GBM cells and fibroblasts.
Collapse
Affiliation(s)
- Dimitra Manou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece; (D.M.); (M.-A.G.)
| | - Maria-Angeliki Golfinopoulou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece; (D.M.); (M.-A.G.)
| | - Sara Naif D. Alharbi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (S.N.D.A.); (H.A.A.); (F.M.A.)
| | - Hind A. Alghamdi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (S.N.D.A.); (H.A.A.); (F.M.A.)
| | - Fatimah Mohammed Alzahrani
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (S.N.D.A.); (H.A.A.); (F.M.A.)
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece; (D.M.); (M.-A.G.)
| |
Collapse
|
7
|
Onciul R, Brehar FM, Toader C, Covache-Busuioc RA, Glavan LA, Bratu BG, Costin HP, Dumitrascu DI, Serban M, Ciurea AV. Deciphering Glioblastoma: Fundamental and Novel Insights into the Biology and Therapeutic Strategies of Gliomas. Curr Issues Mol Biol 2024; 46:2402-2443. [PMID: 38534769 DOI: 10.3390/cimb46030153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/06/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
Gliomas constitute a diverse and complex array of tumors within the central nervous system (CNS), characterized by a wide range of prognostic outcomes and responses to therapeutic interventions. This literature review endeavors to conduct a thorough investigation of gliomas, with a particular emphasis on glioblastoma (GBM), beginning with their classification and epidemiological characteristics, evaluating their relative importance within the CNS tumor spectrum. We examine the immunological context of gliomas, unveiling the intricate immune environment and its ramifications for disease progression and therapeutic strategies. Moreover, we accentuate critical developments in understanding tumor behavior, focusing on recent research breakthroughs in treatment responses and the elucidation of cellular signaling pathways. Analyzing the most novel transcriptomic studies, we investigate the variations in gene expression patterns in glioma cells, assessing the prognostic and therapeutic implications of these genetic alterations. Furthermore, the role of epigenetic modifications in the pathogenesis of gliomas is underscored, suggesting that such changes are fundamental to tumor evolution and possible therapeutic advancements. In the end, this comparative oncological analysis situates GBM within the wider context of neoplasms, delineating both distinct and shared characteristics with other types of tumors.
Collapse
Affiliation(s)
- Razvan Onciul
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Neurosurgery Department, Emergency University Hospital, 050098 Bucharest, Romania
| | - Felix-Mircea Brehar
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurosurgery, Clinical Emergency Hospital "Bagdasar-Arseni", 041915 Bucharest, Romania
| | - Corneliu Toader
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | | | - Luca-Andrei Glavan
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Bogdan-Gabriel Bratu
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Horia Petre Costin
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - David-Ioan Dumitrascu
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Matei Serban
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
8
|
Nafe R, Hattingen E. Cellular Components of the Tumor Environment in Gliomas-What Do We Know Today? Biomedicines 2023; 12:14. [PMID: 38275375 PMCID: PMC10813739 DOI: 10.3390/biomedicines12010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
A generation ago, the molecular properties of tumor cells were the focus of scientific interest in oncology research. Since then, it has become increasingly apparent that the tumor environment (TEM), whose major components are non-neoplastic cell types, is also of utmost importance for our understanding of tumor growth, maintenance and resistance. In this review, we present the current knowledge concerning all cellular components within the TEM in gliomas, focusing on their molecular properties, expression patterns and influence on the biological behavior of gliomas. Insight into the TEM of gliomas has expanded considerably in recent years, including many aspects that previously received only marginal attention, such as the phenomenon of phagocytosis of glioma cells by macrophages and the role of the thyroid-stimulating hormone on glioma growth. We also discuss other topics such as the migration of lymphocytes into the tumor, phenotypic similarities between chemoresistant glioma cells and stem cells, and new clinical approaches with immunotherapies involving the cells of TEM.
Collapse
Affiliation(s)
- Reinhold Nafe
- Department of Neuroradiology, Clinics of Johann Wolfgang Goethe-University, Schleusenweg 2-16, D-60528 Frankfurt am Main, Germany;
| | | |
Collapse
|
9
|
Blanchard R, Adjei I. Engineering the glioblastoma microenvironment with bioactive nanoparticles for effective immunotherapy. RSC Adv 2023; 13:31411-31425. [PMID: 37901257 PMCID: PMC10603567 DOI: 10.1039/d3ra01153d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
While immunotherapies have revolutionized treatment for other cancers, glioblastoma multiforme (GBM) patients have not shown similar positive responses. The limited response to immunotherapies is partly due to the unique challenges associated with the GBM tumor microenvironment (TME), which promotes resistance to immunotherapies, causing many promising therapies to fail. There is, therefore, an urgent need to develop strategies that make the TME immune permissive to promote treatment efficacy. Bioactive nano-delivery systems, in which the nanoparticle, due to its chemical composition, provides the pharmacological function, have recently emerged as an encouraging option for enhancing the efficacy of immunotherapeutics. These systems are designed to overcome immunosuppressive mechanisms in the TME to improve the efficacy of a therapy. This review will discuss different aspects of the TME and how they impede therapy success. Then, we will summarize recent developments in TME-modifying nanotherapeutics and the in vitro models utilized to facilitate these advances.
Collapse
Affiliation(s)
- Ryan Blanchard
- Department of Biomedical Engineering, Texas A&M University TX USA
| | - Isaac Adjei
- Department of Biomedical Engineering, Texas A&M University TX USA
| |
Collapse
|
10
|
Fejza A, Carobolante G, Poletto E, Camicia L, Schinello G, Di Siena E, Ricci G, Mongiat M, Andreuzzi E. The entanglement of extracellular matrix molecules and immune checkpoint inhibitors in cancer: a systematic review of the literature. Front Immunol 2023; 14:1270981. [PMID: 37854588 PMCID: PMC10579931 DOI: 10.3389/fimmu.2023.1270981] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Introduction Immune-checkpoint inhibitors (ICIs) have emerged as a core pillar of cancer therapy as single agents or in combination regimens both in adults and children. Unfortunately, ICIs provide a long-lasting therapeutic effect in only one third of the patients. Thus, the search for predictive biomarkers of responsiveness to ICIs remains an urgent clinical need. The efficacy of ICIs treatments is strongly affected not only by the specific characteristics of cancer cells and the levels of immune checkpoint ligands, but also by other components of the tumor microenvironment, among which the extracellular matrix (ECM) is emerging as key player. With the aim to comprehensively describe the relation between ECM and ICIs' efficacy in cancer patients, the present review systematically evaluated the current literature regarding ECM remodeling in association with immunotherapeutic approaches. Methods This review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines and was registered at the International Prospective Register of Systematic Reviews (PROSPERO, CRD42022351180). PubMed, Web of Science, and Scopus databases were comprehensively searched from inception to January 2023. Titles, abstracts and full text screening was performed to exclude non eligible articles. The risk of bias was assessed using the QUADAS-2 tool. Results After employing relevant MeSH and key terms, we identified a total of 5070 studies. Among them, 2540 duplicates, 1521 reviews or commentaries were found and excluded. Following title and abstract screening, the full text was analyzed, and 47 studies meeting the eligibility criteria were retained. The studies included in this systematic review comprehensively recapitulate the latest observations associating changes of the ECM composition following remodeling with the traits of the tumor immune cell infiltration. The present study provides for the first time a broad view of the tight association between ECM molecules and ICIs efficacy in different tumor types, highlighting the importance of ECM-derived proteolytic products as promising liquid biopsy-based biomarkers to predict the efficacy of ICIs. Conclusion ECM remodeling has an important impact on the immune traits of different tumor types. Increasing evidence pinpoint at ECM-derived molecules as putative biomarkers to identify the patients that would most likely benefit from ICIs treatments. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022351180, identifier CRD42022351180.
Collapse
Affiliation(s)
- Albina Fejza
- Department of Biochemistry, Faculty of Medical Sciences, UBT-Higher Education Institute, Prishtina, Kosovo
| | - Greta Carobolante
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Lucrezia Camicia
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giorgia Schinello
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Emanuele Di Siena
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giuseppe Ricci
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Eva Andreuzzi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
11
|
The Tumor Immune Microenvironment in Primary CNS Neoplasms: A Review of Current Knowledge and Therapeutic Approaches. Int J Mol Sci 2023; 24:ijms24032020. [PMID: 36768342 PMCID: PMC9917056 DOI: 10.3390/ijms24032020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Primary CNS neoplasms are responsible for considerable mortality and morbidity, and many therapies directed at primary brain tumors have proven unsuccessful despite their success in preclinical studies. Recently, the tumor immune microenvironment has emerged as a critical aspect of primary CNS neoplasms that may affect their malignancy, prognosis, and response to therapy across patients and tumor grades. This review covers the tumor microenvironment of various primary CNS neoplasms, with a focus on glioblastoma and meningioma. Additionally, current therapeutic strategies based on elements of the tumor microenvironment, including checkpoint inhibitor therapy and immunotherapeutic vaccines, are discussed.
Collapse
|
12
|
Myeloid cell heterogeneity in the tumor microenvironment and therapeutic implications for childhood central nervous system (CNS) tumors. J Neuroimmunol 2023; 374:578009. [PMID: 36508930 DOI: 10.1016/j.jneuroim.2022.578009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/07/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Central nervous system (CNS) tumors are the most common type of solid tumors in children and the leading cause of cancer deaths in ages 0-14. Recent advances in the field of tumor biology and immunology have underscored the disparate nature of these distinct CNS tumor types. In this review, we briefly introduce pediatric CNS tumors and discuss various components of the TME, with a particular focus on myeloid cells. Although most studies regarding myeloid cells have been done on adult CNS tumors and animal models, we discuss the role of myeloid cell heterogeneity in pediatric CNS tumors and describe how these cells may contribute to tumorigenesis and treatment response. In addition, we present studies within the last 5 years that highlight human CNS tumors, the utility of various murine CNS tumor models, and the latest multi-dimensional tools that can be leveraged to investigate myeloid cell infiltration in young adults and children diagnosed with select CNS tumors.
Collapse
|
13
|
Liu YY, Yao RQ, Long LY, Liu YX, Tao BY, Liu HY, Liu JL, Li Z, Chen L, Yao YM. Worldwide productivity and research trend of publications concerning glioma-associated macrophage/microglia: A bibliometric study. Front Neurol 2022; 13:1047162. [PMID: 36570441 PMCID: PMC9772275 DOI: 10.3389/fneur.2022.1047162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Glioma-associated macrophage/microglia (GAM) represents a key player in shaping a unique glioma ecosystem to facilitate tumor progression and therapeutic resistance. Numerous studies have been published concerning GAM, but no relevant bibliometric study has been performed yet. Our bibliometric study aimed to comprehensively summarize and analyze the global scientific output, research hotspots, and trendy topics of publications on GAM over time. Data on publications on GAM were collected using the Web of Science (WoS). The search date was 16 January 2022, and the publications were collected from 2002 to 2021. Totally, 1,224 articles and reviews were incorporated and analyzed in the current study. It showed that the annual publications concerning GAM kept increasing over the past 20 years. The United States had the largest number of publications and total citations. Holland, Kettenmann, and Gutmann were the top three authors in terms of citation frequency. Neuro-oncology represented the most influential journal in GAM studies, with the highest H-index, total citations, and publication numbers. The paper published by Hambardzumyan in 2016 had the highest local citations. Additionally, the analysis of keywords implied that "prognosis," "tumor microenvironment," and "immunotherapy" might become research hotspots. Furthermore, trendy topics in GAM studies suggested that "immune infiltration," "immune microenvironment," "bioinformatics," "prognosis," and "immunotherapy" deserved additional attention. In conclusion, this bibliometric study comprehensively analyzed the publication trend of GAM studies for the past 20 years, in which the research hotspots and trendy topics were also uncovered. This information offered scholars critical references for conducting in-depth studies on GAM in the future.
Collapse
Affiliation(s)
- Yu-yang Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China,Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Li-yan Long
- Library, Medical School of Chinese PLA, Beijing, China
| | - Yu-xiao Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Bing-Yan Tao
- Medical School of Chinese PLA, Beijing, China,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Hong-yu Liu
- Medical School of Chinese PLA, Beijing, China,Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Jia-lin Liu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ze Li
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Ling Chen
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China,Ling Chen
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China,*Correspondence: Yong-ming Yao
| |
Collapse
|
14
|
van Bree NFHN, Wilhelm M. The Tumor Microenvironment of Medulloblastoma: An Intricate Multicellular Network with Therapeutic Potential. Cancers (Basel) 2022; 14:5009. [PMID: 36291792 PMCID: PMC9599673 DOI: 10.3390/cancers14205009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Medulloblastoma (MB) is a heterogeneous disease in which survival is highly affected by the underlying subgroup-specific characteristics. Although the current treatment modalities have increased the overall survival rates of MB up to 70-80%, MB remains a major cause of cancer-related mortality among children. This indicates that novel therapeutic approaches against MB are needed. New promising treatment options comprise the targeting of cells and components of the tumor microenvironment (TME). The TME of MB consists of an intricate multicellular network of tumor cells, progenitor cells, astrocytes, neurons, supporting stromal cells, microglia, immune cells, extracellular matrix components, and vasculature systems. In this review, we will discuss all the different components of the MB TME and their role in MB initiation, progression, metastasis, and relapse. Additionally, we briefly introduce the effect that age plays on the TME of brain malignancies and discuss the MB subgroup-specific differences in TME components and how all of these variations could affect the progression of MB. Finally, we highlight the TME-directed treatments, in which we will focus on therapies that are being evaluated in clinical trials.
Collapse
Affiliation(s)
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, 17165 Stockholm, Sweden
| |
Collapse
|
15
|
Zhuo S, He G, Chen T, Li X, Liang Y, Wu W, Weng L, Feng J, Gao Z, Yang K. Emerging role of ferroptosis in glioblastoma: Therapeutic opportunities and challenges. Front Mol Biosci 2022; 9:974156. [PMID: 36060242 PMCID: PMC9428609 DOI: 10.3389/fmolb.2022.974156] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant craniocerebral tumor. The treatment of this cancer is difficult due to its high heterogeneity and immunosuppressive microenvironment. Ferroptosis is a newly found non-apoptotic regulatory cell death process that plays a vital role in a variety of brain diseases, including cerebral hemorrhage, neurodegenerative diseases, and primary or metastatic brain tumors. Recent studies have shown that targeting ferroptosis can be an effective strategy to overcome resistance to tumor therapy and immune escape mechanisms. This suggests that combining ferroptosis-based therapies with other treatments may be an effective strategy to improve the treatment of GBM. Here, we critically reviewed existing studies on the effect of ferroptosis on GBM therapies such as chemotherapy, radiotherapy, immunotherapy, and targeted therapy. In particular, this review discussed the potential of ferroptosis inducers to reverse drug resistance and enhance the sensitivity of conventional cancer therapy in combination with ferroptosis. Finally, we highlighted the therapeutic opportunities and challenges facing the clinical application of ferroptosis-based therapies in GBM. The data generated here provide new insights and directions for future research on the significance of ferroptosis-based therapies in GBM.
Collapse
Affiliation(s)
- Shenghua Zhuo
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Guiying He
- Department of Neurology, Shenzhen Sixth People’s Hospital, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Taixue Chen
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiang Li
- Department of Neurology, Shenzhen Sixth People’s Hospital, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Yunheng Liang
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wenkai Wu
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Lingxiao Weng
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jigao Feng
- Department of Neurosurgery, Second Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Kun Yang, ; Zhenzhong Gao, ; Jigao Feng,
| | - Zhenzhong Gao
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Kun Yang, ; Zhenzhong Gao, ; Jigao Feng,
| | - Kun Yang
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Kun Yang, ; Zhenzhong Gao, ; Jigao Feng,
| |
Collapse
|