1
|
Liu W, Liu Y, Li H, Wang S, Chen P, Liu Z, Huo X, Tian J. IGF2BP2 orchestrates global expression and alternative splicing profiles associated with glioblastoma development in U251 cells. Transl Oncol 2025; 51:102177. [PMID: 39515086 PMCID: PMC11582445 DOI: 10.1016/j.tranon.2024.102177] [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: 06/11/2024] [Revised: 09/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma (GBM) is a highly invasive and malignant central nervous system tumor with a median survival duration of 15 months despite multimodal therapy. The insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) has been implicated in various cancers and is known to regulate RNA metabolism and alternative splicing (AS). However, its role in GBM remains unclear. Overexpression of IGF2BP2 led to significant alterations in gene expression, with 472 genes upregulated and 99 downregulated. Gene ontology (GO) analysis indicated enrichment in immune-related biological processes. Notably, IGF2BP2 was found to regulate AS events, with 1372 regulated AS genes (RASGs) and 2096 significantly distinct ASEs identified. Furthermore, IGF2BP2 selectively bound to 3' and 5' untranslated regions (UTRs) via GG[AU]C motifs, and IFIH1 was identified as a direct binding partner and upregulated gene upon IGF2BP2 overexpression. Functional enrichment analysis suggested that IGF2BP2 influences pathways related to RNA splicing and immune responses. Our findings demonstrate that IGF2BP2 plays a crucial role in GBM by modulating the transcriptome and AS events. The upregulation of immune-related genes and the regulation of AS by IGF2BP2 highlight its potential as a therapeutic target in GBM, particularly for immunotherapy. The study provides a foundation for further investigation into the molecular mechanisms of IGF2BP2 in GBM and its implications for cancer treatment.
Collapse
Affiliation(s)
- Wenqing Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yan Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China; Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, Ningxia, China; Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haoyuan Li
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China; Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, Ningxia, China; Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shixiong Wang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China; Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Pengfei Chen
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhongtao Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xianhao Huo
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Jihui Tian
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
2
|
Zambom-Ferraresi F, Zambom-Ferraresi F, Izco M, Martínez-Velilla N, Extramiana L, Fernández-Irigoyen J, Santamaría E, Llamas-Urbano A, Hanaee Y, Martinez-Moreno JM, Barbarroja N, Pérez-Sánchez C. Monitoring Cerebrospinal Fluid Inflammatory Mediators by Olink Target 48 Cytokine Panel. Methods Mol Biol 2025; 2914:51-64. [PMID: 40167910 DOI: 10.1007/978-1-0716-4462-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The diagnosis and monitoring of neurological disorders are challenging due to their complexity and the difficulty of obtaining samples of damaged tissue while the patient is alive. Cerebrospinal fluid (CSF) is an excellent biofluid for conducting diagnostic and monitoring studies because it is in direct contact with the brain, allowing for the analysis of secreted proteins and peptides associated with various neurological disorders.The Proximity Extension Assay, which combines antibodies with oligonucleotides to detect proteins via qPCR, is revolutionizing the field of biomarker identification. The Olink Target 48 Cytokine Panel is an optimal tool for evaluating the inflammatory profile of CSF samples, requiring only one microliter. The workflow involves incubation with antibody-linked oligonucleotides, extension, amplification through a PCR assay, and detection using microfluidic qPCR, enabling precise and accurate quantification of cytokine levels in pg/mL. This approach offers valuable insights into inflammatory processes within the CSF, serving as a robust tool for studying and understanding inflammation in neurological contexts.
Collapse
Affiliation(s)
- Fabricio Zambom-Ferraresi
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Fabiola Zambom-Ferraresi
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Maite Izco
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Nicolás Martínez-Velilla
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Leire Extramiana
- Clinical Neuroproteomics Laboratory, Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Laboratory, Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Laboratory, Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | | | - Yasin Hanaee
- Cobiomic Bioscience. EBT University of Cordoba/IMIBIC, Cordoba, Spain
| | | | - Nuria Barbarroja
- Cobiomic Bioscience. EBT University of Cordoba/IMIBIC, Cordoba, Spain
- Rheumatology Unit/Reina Sofia Hospital/University of Cordoba/IMIBIC, Cordoba, Spain
| | - Carlos Pérez-Sánchez
- Cobiomic Bioscience. EBT University of Cordoba/IMIBIC, Cordoba, Spain.
- Rheumatology Unit/Reina Sofia Hospital/University of Cordoba/IMIBIC, Cordoba, Spain.
| |
Collapse
|
3
|
Altinoz MA, Yilmaz A, Taghizadehghalehjoughi A, Genc S, Yeni Y, Gecili I, Hacimuftuoglu A. Ulipristal-temozolomide-hydroxyurea combination for glioblastoma: in-vitro studies. J Neurosurg Sci 2024; 68:468-481. [PMID: 35766205 DOI: 10.23736/s0390-5616.22.05718-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a brain malignancy with worst survival. Low dose progesterone stimulates GBM growth, while progesterone receptor (PR)-antagonist mifepristone was shown to reduce growth and to enhance temozolomide sensitivity in GBM cells. Mifepristone is not available in all countries due to ethical reasons and may cause adrenal insufficiency and pelvic infections. Ulipristal is also a PR-antagonist used in treatment of uterine leiomyomas with higher biosafety. Ulipristal is demonstrated to suppress growth of breast cancer, yet it is not tested as yet whether it can also block growth and sensitize to temozolomide in glioblastoma as it was previously shown with mifepristone. Our first aim was to detect whether ulipristal exerts antiproliferative and chemotherapy-sensitizing effects in glioblastoma. Hydroxyurea inhibits DNA replication via blocking ribonucleotide reductase (RR) and it was demonstrated to increase temozolomide antineoplasticity in GBM. Progesterone receptor-activation in the uterus enhances RR transcription. Hence, we have hypothesized that PR-inactivation with ulipristal would further enhance hydroxyurea antineoplasticity by shutting down DNA synthesis mechanisms through further suppression of RR. Lastly, there exists no study as yet whether ulipristal, hydroxyurea and temozolomide could exert ternary antineoplastic efficacy, which was our last aim to define. METHODS To reveal interactions between ulipristal, hydroxyurea and temozolomide, we treated human U251 GBM cell line with these agents alone and in combination and measured cell proliferation, total antioxidant capacity (TAC) and total oxidant status (TOS) in conditioned medium and cellular cytokine gene expressions. RESULTS All agents significantly reduced cell proliferation significantly, yet the most significant decrease of GBM cells occurred with the triple drug combination at the 96th hour. All agents significantly decreased TAC and increased TOS in culture media, which was mostly relevant for the triple combination at the 96th hour. All these three agents tend to reduce the expression of immunosuppressive and/or GBM-growth stimulating cytokines TGF-β, IL-10 and IL-17 while increasing the expression of GBM-growth suppressing cytokine IL-23. CONCLUSIONS Reproposal of these agents in treatment of GBM would be a plausible approach if future studies prove their efficacy.
Collapse
Affiliation(s)
- Meric A Altinoz
- Department of Biochemistry, Acibadem University, Istanbul, Türkiye -
| | - Aysegul Yilmaz
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | - Ali Taghizadehghalehjoughi
- Department of Veterinary Pharmacology and Toxicology, Veterinary Medicine, Ataturk University, Erzurum, Türkiye
| | - Sidika Genc
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | - Yesim Yeni
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | - Ibrahim Gecili
- Department of Medical Pharmacology, Ataturk University, Erzurum, Türkiye
| | | |
Collapse
|
4
|
Boccalatte F, Mina R, Aroldi A, Leone S, Suryadevara CM, Placantonakis DG, Bruno B. Advances and Hurdles in CAR T Cell Immune Therapy for Solid Tumors. Cancers (Basel) 2022; 14:5108. [PMID: 36291891 PMCID: PMC9600451 DOI: 10.3390/cancers14205108] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells in solid tumors have so far yielded limited results, in terms of therapeutic effects, as compared to the dramatic results observed for hematological malignancies. Many factors involve both the tumor cells and the microenvironment. The lack of specific target antigens and severe, potentially fatal, toxicities caused by on-target off-tumor toxicities constitute major hurdles. Furthermore, the tumor microenvironment is usually characterized by chronic inflammation, the presence of immunosuppressive molecules, and immune cells that can reduce CAR T cell efficacy and facilitate antigen escape. Nonetheless, solid tumors are under investigation as possible targets despite their complexity, which represents a significant challenge. In preclinical mouse models, CAR T cells are able to efficiently recognize and kill several tumor xenografts. Overall, in the next few years, there will be intensive research into optimizing novel cell therapies to improve their effector functions and keep untoward effects in check. In this review, we provide an update on the state-of-the-art CAR T cell therapies in solid tumors, focusing on the preclinical studies and preliminary clinical findings aimed at developing optimal strategies to reduce toxicity and improve efficacy.
Collapse
Affiliation(s)
- Francesco Boccalatte
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Roberto Mina
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, University of Torino, 10126 Torino, TO, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, TO, Italy
| | - Andrea Aroldi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, MB, Italy
| | - Sarah Leone
- Department of Population Health, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Carter M. Suryadevara
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitris G. Placantonakis
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
- Brain and Spine Tumor Center/Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Benedetto Bruno
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, University of Torino, 10126 Torino, TO, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, TO, Italy
| |
Collapse
|
5
|
Singh M, Raghav A, Gautam KA. Role of the circulatory interleukin-6 in the pathogenesis of gliomas: A systematic review. World J Methodol 2022; 12:428-437. [PMID: 36186749 PMCID: PMC9516551 DOI: 10.5662/wjm.v12.i5.428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/01/2022] [Accepted: 07/25/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Glioma is the most common primary tumor in the brain originating from glial cells. In spite of extensive research, the overall survival rate is not enhanced. A number of published articles observed differentially circulating levels of cytokines in glioma. Interleukin-6 (IL-6) protein coded by IL-6 gene is regulated by the immune system and it has been found to have a significant role in progression and apoptosis resistance of glioma.
AIM To review the role of circulatory IL-6 in the development and progression of glioma and its utility as a biomarker.
METHODS Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines were applied to filter the relevant studies based on inclusion and exclusion criteria. We used a combination of keywords and the Reference Citation Analysis (RCA) tool to search the potential studies and performed data extraction from selected studies.
RESULTS The published results were inconsistent; however, most studies showed a significantly higher IL-6 level in glioma cases as compared to controls. Comparative IL-6 level among the different grades of glioma showed a higher level with low-grade gliomas and lower level with high-grade gliomas.
CONCLUSION IL-6 level significantly differed between cases and controls, and among different cancer stages, which shows its potential as a diagnostic and prognostic marker.
Collapse
Affiliation(s)
- Manish Singh
- Department of Neurosurgery, GSVM Medical College, Kanpur 208001, India
| | - Alok Raghav
- Department of Neurosurgery, GSVM Medical College, Kanpur 208001, India
| | - Kirti Amresh Gautam
- Department of Basic and Applied Science, GD Goenka University, Gurugram 122103, Haryana, India
| |
Collapse
|
6
|
Petrovic M, Porcello A, Tankov S, Majchrzak O, Kiening M, Laingoniaina AC, Jbilou T, Walker PR, Borchard G, Jordan O. Synthesis, Formulation and Characterization of Immunotherapeutic Glycosylated Dendrimer/cGAMP Complexes for CD206 Targeted Delivery to M2 Macrophages in Cold Tumors. Pharmaceutics 2022; 14:pharmaceutics14091883. [PMID: 36145631 PMCID: PMC9503622 DOI: 10.3390/pharmaceutics14091883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Anti-tumor responses can be achieved via the stimulation of the immune system, a therapeutic approach called cancer immunotherapy. Many solid tumor types are characterized by the presence of immune-suppressive tumor-associated macrophage (TAMs) cells within the tumor microenvironment (TME). Moreover, TAM infiltration is strongly associated with poor survival in solid cancer patients and hence a low responsiveness to cancer immunotherapy. Therefore, 2′3′ Cyclic GMP-AMP (2′3′ cGAMP) was employed for its ability to shift macrophages from pro-tumoral M2-like macrophages (TAM) to anti-tumoral M1. However, cGAMP transfection within macrophages is limited by the molecule’s negative charge, poor stability and lack of targeting. To circumvent these barriers, we designed nanocarriers based on poly(amidoamine) dendrimers (PAMAM) grafted with D-glucuronic acid (Glu) for M2 mannose-mediated endocytosis. Two carriers were synthesized based on different dendrimers and complexed with cGAMP at different ratios. Orthogonal techniques were employed for synthesis (NMR, ninhydrin, and gravimetry), size (DLS, NTA, and AF4-DLS), charge (DLS and NTA), complexation (HPLC-UV and AF4-UV) and biocompatibility and toxicity (primary cells and hen egg chorioallantoic membrane model) evaluations in order to evaluate the best cGAMP carrier. The best formulation was selected for its low toxicity, biocompatibility, monodispersed distribution, affinity towards CD206 and ability to increase M1 (STAT1 and NOS2) and decrease M2 marker (MRC1) expression in macrophages.
Collapse
Affiliation(s)
- Marija Petrovic
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Alexandre Porcello
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Stoyan Tankov
- Translational Research Centre in Oncohaematology, University of Geneva, 1206 Geneva, Switzerland
| | - Oliwia Majchrzak
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Martin Kiening
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Annick Clara Laingoniaina
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Tayeb Jbilou
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Paul R. Walker
- Translational Research Centre in Oncohaematology, University of Geneva, 1206 Geneva, Switzerland
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1206 Geneva, Switzerland
- Correspondence: ; Tel.: +41-223796586
| |
Collapse
|
7
|
Liang J, Liu F, Yang Y, Li X, Cai G, Cao J, Zhang B. Diagnostic and prognostic utility of eIF6 in glioblastoma: a study based on TCGA and CGGA databases. Am J Transl Res 2022; 14:5040-5049. [PMID: 35958479 PMCID: PMC9360856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Among various glioma types, glioblastoma multiforme (GBM) is one of those with the highest malignancy. Although overexpression of eukaryotic translation initiation factor 6 (eIF6), a factor that regulates protein translation initiation, is believed to promote tumor development, its function and potential molecular mechanisms in glioma progression remain uncharacterized. Consequently, we evaluated its diagnostic and prognostic utility in GBM patients. METHODS Sample data from two databases, The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), were utilized to investigate the role of eIF6 as well as its mechanism of action in gliomas. We analyzed eIF6 expression in normal tissues as well as cancerous samples of different stages of glioma. The diagnostic and prognostic value of eIF6 were analyzed using the Receiver Operating Characteristic Curve (ROC) and Kaplan-Meier analysis, respectively. Furthermore, its underlying molecular mechanism in GBM was further revealed by gene set enrichment analysis (GSEA). RESULTS Transcriptome data analyses of the two databases showed that eIF6 was upregulated in glioma tissues compared with normal counterparts. eIF6 was at high levels in WHO grade IV gliomas versus grade II and III gliomas (P<0.05). In addition, eIF6 was highly expressed in elderly and Asian glioma patients. Furthermore, eIF6 expression was found to be lower in isocitrate dehydrogenase (IDH)-mutated tumors. Patients with high eIF6 level presented shorter overall survival than cases with low eIF6 level (P<0.05), and eIF6 had favorable accuracy in predicting the prognosis of glioma patients. GSEA revealed that high eIF6 expression was mainly concentrated in cell cycle and DNA repair related pathways. CONCLUSIONS eIF6 is highly expressed in gliomas and positively associated with the degree of malignancy. Patients with high eIF6 expression present poor survival. Therefore, eIF6 has the potential to be a diagnostic biomarker and a potential therapeutic target for glioma development and GBM.
Collapse
Affiliation(s)
- Jian Liang
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Fengyu Liu
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Yaoqiang Yang
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Xing Li
- School of Medicine, Southern University of Science and TechnologyShenzhen 518055, Guangdong, China
| | - Guangmou Cai
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Jianxuan Cao
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Bo Zhang
- Department of Neurosurgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| |
Collapse
|
8
|
Wang G, Wang J, Niu C, Zhao Y, Wu P. Neutrophils: New Critical Regulators of Glioma. Front Immunol 2022; 13:927233. [PMID: 35860278 PMCID: PMC9289230 DOI: 10.3389/fimmu.2022.927233] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/06/2022] [Indexed: 11/22/2022] Open
Abstract
In cancer, neutrophils are an important part of the tumour microenvironment (TME). Previous studies have shown that circulating and infiltrating neutrophils are associated with malignant progression and immunosuppression in gliomas. However, recent studies have shown that neutrophils have an antitumour effect. In this review, we focus on the functional roles of neutrophils in the circulation and tumour sites in patients with glioma. The mechanisms of neutrophil recruitment, immunosuppression and the differentiation of neutrophils are discussed. Finally, the potential of neutrophils as clinical biomarkers and therapeutic targets is highlighted. This review can help us gain a deeper and systematic understanding of the role of neutrophils, and provide new insights for treatment in gliomas.
Collapse
Affiliation(s)
- Guanyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinpeng Wang
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaoshi Niu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Stereotactic Neurosurgical Institute, Hefei, China
- Anhui Province Key Laboratory of Brain Function and Brain Disease, Hefei, China
- Anhui Provincial Clinical Research Center for Neurosurgical Disease, Hefei, China
| | - Yan Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengfei Wu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Stereotactic Neurosurgical Institute, Hefei, China
- Anhui Province Key Laboratory of Brain Function and Brain Disease, Hefei, China
- Anhui Provincial Clinical Research Center for Neurosurgical Disease, Hefei, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| |
Collapse
|
9
|
Chitadze G, Kabelitz D. Immune surveillance in glioblastoma: role of the NKG2D system and novel cell-based therapeutic approaches. Scand J Immunol 2022; 96:e13201. [PMID: 35778892 DOI: 10.1111/sji.13201] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/27/2022]
Abstract
Glioblastoma, formerly known as Glioblastoma multiforme (GBM) is the most frequent and most aggressive brain tumor in adults. The brain is an immunopriviledged organ and the blood brain barrier shields the brain from immune surveillance. In this review we discuss the composition of the immunosuppressive tumor micromilieu and potential immune escape mechanisms in GBM. In this respect, we focus on the role of the NKG2D receptor/ligand system. NKG2D ligands are frequently expressed on GBM tumor cells and can activate NKG2D-expressing killer cells including NK cells and γδ T cells. Soluble NKG2D ligands, however, contribute to tumor escape from immunological attack. We also discuss the current immunotherapeutic strategies to improve the survival of GBM patients. Such approaches include the modulation of the NKG2D receptor/ligand system, the application of checkpoint inhibitors, the adoptive transfer of ex vivo expanded and/or modified immune cells, or the application of antibodies and antibody constructs to target cytotoxic effector cells in vivo. In view of the multitude of pursued strategies, there is hope for improved overall survival of GBM patients in the future.
Collapse
Affiliation(s)
- Guranda Chitadze
- Unit for Hematological Diagnostics, Department of Internal Medicine II
| | - Dieter Kabelitz
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| |
Collapse
|
10
|
Liu B, Ji Q, Cheng Y, Liu M, Zhang B, Mei Q, Liu D, Zhou S. Biomimetic GBM-targeted drug delivery system boosting ferroptosis for immunotherapy of orthotopic drug-resistant GBM. J Nanobiotechnology 2022; 20:161. [PMID: 35351131 PMCID: PMC8962245 DOI: 10.1186/s12951-022-01360-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/08/2022] [Indexed: 01/13/2023] Open
Abstract
Background Clinical studies have shown that the efficacy of programmed cell death receptor-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors on glioblastoma (GBM) is much lower than what is expected because of the low immunogenicity of GBM. Ferroptosis of cancer cells can induce the maturation of dendritic cells (DC cells) and increase the activity of T cell. The activated T cells release IFN-γ, which subsequently induces the ferroptosis of cancer cells. Thus, the aim of this paper is to set up a new GBM-targeted drug delivery system (Fe3O4-siPD-L1@M-BV2) to boost ferroptosis for immunotherapy of drug-resistant GBM. Results Fe3O4-siPD-L1@M-BV2 significantly increased the accumulation of siPD-L1 and Fe2+ in orthotopic drug-resistant GBM tissue in mice. Fe3O4-siPD-L1@M-BV2 markedly decreased the protein expression of PD-L1 and increased the ratio between effector T cells and regulatory T cells in orthotopic drug-resistant GBM tissue. Moreover, Fe3O4-siPD-L1@M-BV2 induced ferroptosis of GBM cells and maturation of DC cell, and it also increased the ratio between M1-type microglia and M2-type microglia in orthotopic drug-resistant GBM tissue. Finally, the growth of orthotopic drug-resistant GBM in mice was significantly inhibited by Fe3O4-siPD-L1@M-BV2. Conclusion The mutual cascade amplification effect between ferroptosis and immune reactivation induced by Fe3O4-siPD-L1@M-BV2 significantly inhibited the growth of orthotopic drug-resistant GBM and prolonged the survival time of orthotopic drug-resistant GBM mice. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01360-6.
Collapse
Affiliation(s)
- Bao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qibing Mei
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China. .,Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
11
|
Challenges and Opportunities for Immunotherapeutic Intervention against Myeloid Immunosuppression in Glioblastoma. J Clin Med 2022; 11:jcm11041069. [PMID: 35207340 PMCID: PMC8880446 DOI: 10.3390/jcm11041069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common and deadly brain cancer, exemplifies the paradigm that cancers grow with help from an immunosuppressive tumor microenvironment (TME). In general, TME includes a large contribution from various myeloid lineage-derived cell types, including (in the brain) altered pathogenic microglia as well as monocyte-macrophages (Macs), myeloid-derived suppressor cells (MDSC) and dendritic cell (DC) populations. Each can have protective roles, but has, by definition, been coopted by the tumor in patients with progressive disease. However, evidence demonstrates that myeloid immunosuppressive activities can be reversed in different ways, leading to enthusiasm for this therapeutic approach, both alone and in combination with potentially synergistic immunotherapeutic and other strategies. Here, we review the current understanding of myeloid cell immunosuppression of anti-tumor responses as well as potential targets, challenges, and developing means to reverse immunosuppression with various therapeutics and their status. Targets include myeloid cell colony stimulating factors (CSFs), insulin-like growth factor 1 (IGF1), several cytokines and chemokines, as well as CD40 activation and COX2 inhibition. Approaches in clinical development include antibodies, antisense RNA-based drugs, cell-based combinations, polarizing cytokines, and utilizing Macs as a platform for Chimeric Antigen Receptors (CAR)-based tumor targeting, like with CAR-T cells. To date, promising clinical results have been reported with several of these approaches.
Collapse
|
12
|
Dapash M, Hou D, Castro B, Lee-Chang C, Lesniak MS. The Interplay between Glioblastoma and Its Microenvironment. Cells 2021; 10:2257. [PMID: 34571905 PMCID: PMC8469987 DOI: 10.3390/cells10092257] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 01/05/2023] Open
Abstract
GBM is the most common primary brain tumor in adults, and the aggressive nature of this tumor contributes to its extremely poor prognosis. Over the years, the heterogeneous and adaptive nature of GBM has been highlighted as a major contributor to the poor efficacy of many treatments including various immunotherapies. The major challenge lies in understanding and manipulating the complex interplay among the different components within the tumor microenvironment (TME). This interplay varies not only by the type of cells interacting but also by their spatial distribution with the TME. This review highlights the various immune and non-immune components of the tumor microenvironment and their consequences of the efficacy of immunotherapies. Understanding the independent and interdependent aspects of the various sub-populations encapsulated by the immune and non-immune components will allow for more targeted therapies. Meanwhile, understanding how the TME creates and responds to different environmental pressures such as hypoxia may allow for other multimodal approaches in the treatment of GBM. Ultimately, a better understanding of the GBM TME will aid in the development and advancement of more effective treatments and in improving patient outcomes.
Collapse
Affiliation(s)
- Mark Dapash
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.D.); (D.H.); (B.C.)
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - David Hou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.D.); (D.H.); (B.C.)
| | - Brandyn Castro
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.D.); (D.H.); (B.C.)
- Department of Neurosurgery, University of Chicago, Chicago, IL 60637, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.D.); (D.H.); (B.C.)
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.D.); (D.H.); (B.C.)
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
13
|
Arcos-Montoya D, Wegman-Ostrosky T, Mejía-Pérez S, De la Fuente-Granada M, Camacho-Arroyo I, García-Carrancá A, Velasco-Velázquez MA, Manjarrez-Marmolejo J, González-Arenas A. Progesterone Receptor Together with PKCα Expression as Prognostic Factors for Astrocytomas Malignancy. Onco Targets Ther 2021; 14:3757-3768. [PMID: 34168461 PMCID: PMC8217595 DOI: 10.2147/ott.s280314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/20/2020] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Astrocytomas are the most common and aggressive primary brain tumors, and they are classified according to the degree of malignancy on a scale of I to IV, in which grade I is the least malignant and grade IV the highest. Many factors are related to astrocytomas progression as progesterone receptor (PR), whose transcriptional activity could be regulated by phosphorylation by protein kinase C alpha (PKCα) at the residue Ser400. Our aim was to investigate if PR phosphorylation together with PKCα expression could be used as a prognostic factor for astrocytomas malignancy. METHODS By immunofluorescence, we detected the content of PKCα, PR and its phosphorylation at Ser400 in 46 biopsies from Mexican patients with different astrocytoma malignancy grades; by bioinformatic tools using TCGA data, we evaluated the expression of PR and PKCα mRNA according to astrocytoma malignancy grades. For all statistical analyses, significance was p<0.05. RESULTS We detected a positive correlation between the tumor grade and the content of PKCα, PR and its phosphorylation at Ser400, as well as the intracellular colocalization of these proteins. Interestingly, using an in silico assay, we found that the PR and PKCα expression at mRNA level has an inverse ratio with astrocytomas tumor grade. DISCUSSION These results indicate that PR and its phosphorylation at Ser400 site, as well as PKCα and their colocalization, could be considered as possible malignancy biomarkers for astrocytomas grades I-IV.
Collapse
Affiliation(s)
- Denisse Arcos-Montoya
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Talia Wegman-Ostrosky
- Dirección de Investigación, Instituto Nacional Cancerología, Ciudad de México, México
- División de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Sonia Mejía-Pérez
- Subdirección de Neurocirugía, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| | - Marisol De la Fuente-Granada
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alejandro García-Carrancá
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México e Instituto Nacional de Cancerología, Ciudad de México, México
| | - Marco A Velasco-Velázquez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Joaquín Manjarrez-Marmolejo
- Laboratorio de Fisiología de la Formación Reticular, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
14
|
Qiu R, Zhong Y, Li Q, Li Y, Fan H. Metabolic Remodeling in Glioma Immune Microenvironment: Intercellular Interactions Distinct From Peripheral Tumors. Front Cell Dev Biol 2021; 9:693215. [PMID: 34211978 PMCID: PMC8239469 DOI: 10.3389/fcell.2021.693215] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 05/19/2021] [Indexed: 01/29/2023] Open
Abstract
During metabolic reprogramming, glioma cells and their initiating cells efficiently utilized carbohydrates, lipids and amino acids in the hypoxic lesions, which not only ensured sufficient energy for rapid growth and improved the migration to normal brain tissues, but also altered the role of immune cells in tumor microenvironment. Glioma cells secreted interferential metabolites or depriving nutrients to injure the tumor recognition, phagocytosis and lysis of glioma-associated microglia/macrophages (GAMs), cytotoxic T lymphocytes, natural killer cells and dendritic cells, promoted the expansion and infiltration of immunosuppressive regulatory T cells and myeloid-derived suppressor cells, and conferred immune silencing phenotypes on GAMs and dendritic cells. The overexpressed metabolic enzymes also increased the secretion of chemokines to attract neutrophils, regulatory T cells, GAMs, and dendritic cells, while weakening the recruitment of cytotoxic T lymphocytes and natural killer cells, which activated anti-inflammatory and tolerant mechanisms and hindered anti-tumor responses. Therefore, brain-targeted metabolic therapy may improve glioma immunity. This review will clarify the metabolic properties of glioma cells and their interactions with tumor microenvironment immunity, and discuss the application strategies of metabolic therapy in glioma immune silence and escape.
Collapse
Affiliation(s)
- Runze Qiu
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Zhong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qingquan Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingbin Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongwei Fan
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Musashi-1 Regulates MIF1-Mediated M2 Macrophage Polarization in Promoting Glioblastoma Progression. Cancers (Basel) 2021; 13:cancers13081799. [PMID: 33918794 PMCID: PMC8069545 DOI: 10.3390/cancers13081799] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/17/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) is the most lethal type of brain cancer. It is well known that the malignancy of cancers is dependent not only on the oncogenic properties of the tumor cells, but also on the composition of the tumor microenvironment, which includes macrophages of the immune system. The prevalence of M2 type macrophages usually promotes tumor progression as opposed to tumor-suppressing function of M1 type macrophages. In our previous studies, we identified Musashi-1 (MSI1) RNA-binding protein as a principal oncogenic factor in GBM. In this study, in a pursuit of finding secreted factors that may alter tumor microenvironment in GBM, we identified MIF1 cytokine to be positively regulated by MSI1. Moreover, we found that MSI1-mediated MIF1 secretion promotes differentiation of macrophages into pro-oncogenic M2 phenotype. The oncogenic role of MSI1/MIF1/M2 macrophage regulatory axis was also confirmed in GBM mouse models, which makes it a promising target for novel drug discovery. Abstract Glioblastoma (GBM) is the most malignant brain tumor which is characterized by high proliferation and migration capacity. The poor survival rate has been attributed to limitations of the current standard therapies. The search for novel biological targets that can effectively hamper tumor progression remains extremely challenging. Previous studies indicated that tumor-associated macrophages (TAMs) are the abundant elements in the tumor microenvironment that are closely implicated in glioma progression and tumor pathogenesis. M2 type TAMs are immunosuppressive and promote GBM proliferation. RNA-binding protein Musashi-1 (MSI1) has recently been identified as a marker of neural stem/progenitor cells, and its high expression has been shown to correlate with the growth of GBM. Nevertheless, the relationship between MSI1 and TAMs in GBM is still unknown. Thus, in our present study, we aimed to investigate the molecular interplay between MSI1 and TAMs in contributing to GBM tumorigenesis. Our data revealed that the secretion of macrophage inhibitory factor 1 (MIF1) is significantly upregulated by MSI1 overexpression in vitro. Importantly, M2 surface markers of THP-1-derived macrophages were induced by recombinant MIF1 and reduced by using MIF1 inhibitor (S,R)-3-(4-hHydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (ISO-1). Furthermore, GBM tumor model data suggested that the tumor growth, MIF1 expression and M2 macrophage population were significantly downregulated when MSI1 expression was silenced in vivo. Collectively, our findings identified a novel role of MSI1 in the secretion of MIF1 and the consequent polarization of macrophages into the M2 phenotype in promoting GBM tumor progression.
Collapse
|
16
|
Decipher the Glioblastoma Microenvironment: The First Milestone for New Groundbreaking Therapeutic Strategies. Genes (Basel) 2021; 12:genes12030445. [PMID: 33804731 PMCID: PMC8003887 DOI: 10.3390/genes12030445] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumour in adults. Despite the combination of novel therapeutical approaches, it remains a deadly malignancy with an abysmal prognosis. GBM is a polymorphic tumour from both molecular and histological points of view. It consists of different malignant cells and various stromal cells, contributing to tumour initiation, progression, and treatment response. GBM’s microenvironment is multifaceted and is made up of soluble factors, extracellular matrix components, tissue-resident cell types (e.g., neurons, astrocytes, endothelial cells, pericytes, and fibroblasts) together with resident (e.g., microglia) or recruited (e.g., bone marrow-derived macrophages) immune cells. These latter constitute the so-called immune microenvironment, accounting for a substantial GBM’s tumour volume. Despite the abundance of immune cells, an intense state of tumour immunosuppression is promoted and developed; this represents the significant challenge for cancer cells’ immune-mediated destruction. Though literature data suggest that distinct GBM’s subtypes harbour differences in their microenvironment, its role in treatment response remains obscure. However, an in-depth investigation of GBM’s microenvironment may lead to novel therapeutic opportunities to improve patients’ outcomes. This review will elucidate the GBM’s microenvironment composition, highlighting the current state of the art in immunotherapy approaches. We will focus on novel strategies of active and passive immunotherapies, including vaccination, gene therapy, checkpoint blockade, and adoptive T-cell therapies.
Collapse
|
17
|
Kazim SF, Martinez E, Hough TJ, Spangler BQ, Bowers CA, Chohan MO. The Survival Benefit of Postoperative Bacterial Infections in Patients With Glioblastoma Multiforme: Myth or Reality? Front Neurol 2021; 12:615593. [PMID: 33613432 PMCID: PMC7894197 DOI: 10.3389/fneur.2021.615593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/14/2021] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma multiforme (GBM), the most common malignant brain tumor, universally carries a poor prognosis. Despite aggressive multimodality treatment, the median survival is ~18-20 months, depending on molecular subgroups. A long history of observations suggests antitumor effects of bacterial infections against malignant tumors. The present review summarizes and critically analyzes the clinical data providing evidence for or against the survival benefit of post-operative bacterial infections in GBM patients. Furthermore, we explore the probable underlying mechanism(s) from basic science studies on the topic. There are plausible explanations from immunobiology for the mechanism of the "favorable effect" of bacterial infections in GBM patients. However, available clinical literature does not provide a definitive association between postoperative bacterial infection and prolonged survival in GBM patients. The presently available, single-/multi-center and national database retrospective case-control studies on the topic provide conflicting results. A prospective randomized study on the subject is clearly not possible. Immunobiology literature supports development of genetically modified bacteria as part of multimodal regimen against GBM.
Collapse
Affiliation(s)
- Syed Faraz Kazim
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, United States
| | - Erick Martinez
- School of Medicine, New York Medical College (NYMC), Valhalla, NY, United States
| | - Tyler J Hough
- School of Medicine, University of New Mexico (UNM), Albuquerque, NM, United States
| | - Benjamin Q Spangler
- School of Medicine, University of New Mexico (UNM), Albuquerque, NM, United States
| | - Christian A Bowers
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, United States
| | - Muhammad Omar Chohan
- Department of Neurosurgery, University of Mississippi Medical Center (UMMC), Jackson, MS, United States
| |
Collapse
|
18
|
Zhao WJ, Ou GY, Lin WW. Integrative Analysis of Neuregulin Family Members-Related Tumor Microenvironment for Predicting the Prognosis in Gliomas. Front Immunol 2021; 12:682415. [PMID: 34054873 PMCID: PMC8155525 DOI: 10.3389/fimmu.2021.682415] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/28/2021] [Indexed: 02/05/2023] Open
Abstract
Gliomas, including brain lower grade glioma (LGG) and glioblastoma multiforme (GBM), are the most common primary brain tumors in the central nervous system. Neuregulin (NRG) family proteins belong to the epidermal growth factor (EGF) family of extracellular ligands and they play an essential role in both the central and peripheral nervous systems. However, roles of NRGs in gliomas, especially their effects on prognosis, still remain to be elucidated. In this study, we obtained raw counts of RNA-sequencing data and corresponding clinical information from 510 LGG and 153 GBM samples from The Cancer Genome Atlas (TCGA) database. We analyzed the association of NRG1-4 expression levels with tumor immune microenvironment in LGG and GBM. GSVA (Gene Set Variation Analysis) was performed to determine the prognostic difference of NRGs gene set between LGG and GBM. ROC (receiver operating characteristic) curve and the nomogram model were constructed to estimate the prognostic value of NRGs in LGG and GBM. The results demonstrated that NRG1-4 were differentially expressed in LGG and GBM in comparison to normal tissue. Immune score analysis revealed that NRG1-4 were significantly related to the tumor immune microenvironment and remarkably correlated with immune cell infiltration. The investigation of roles of m6A (N6-methyladenosine, m6A)-related genes in gliomas revealed that NRGs were prominently involved in m6A RNA modification. GSVA score showed that NRG family members are more associated with prognosis in LGG compared with GBM. Prognostic analysis showed that NRG3 and NRG1 can serve as potential independent biomarkers in LGG and GBM, respectively. Moreover, GDSC drug sensitivity analysis revealed that NRG1 was more correlated with drug response compared with other NRG subtypes. Based on these public databases, we preliminarily identified the relationship between NRG family members and tumor immune microenvironment, and the prognostic value of NRGs in gliomas. In conclusion, our study provides comprehensive roles of NRG family members in gliomas, supporting modulation of NRG signaling in the management of glioma.
Collapse
Affiliation(s)
- Wei-jiang Zhao
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- *Correspondence: Wei-jiang Zhao, ; Guan-yong Ou,
| | - Guan-yong Ou
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- *Correspondence: Wei-jiang Zhao, ; Guan-yong Ou,
| | - Wen-wen Lin
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| |
Collapse
|
19
|
Molecular Characterization of Temozolomide-Treated and Non Temozolomide-Treated Glioblastoma Cells Released Extracellular Vesicles and Their Role in the Macrophage Response. Int J Mol Sci 2020; 21:ijms21218353. [PMID: 33171763 PMCID: PMC7664451 DOI: 10.3390/ijms21218353] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are widely investigated in glioblastoma multiforme (GBM) for their involvement in regulating GBM pathobiology as well as for their use as potential biomarkers. EVs, through cell-to-cell communication, can deliver proteins, nucleic acids, and lipids that are able to reprogram tumor-associated macrophages (TAMs). This research is aimed to concentrate, characterize, and identify molecular markers of EVs subtypes released by temozolomide (TMZ)-treated and non TMZ-treated four diverse GBM cells. Morphology, size distribution, and quantity of small (sEVs) and large (lEVs) vesicles were analyzed by cryo-TEM. Quality and quantity of EVs surface markers were evaluated, having been obtained by Western blotting. GBM cells shed a large amount of EVs, showing a cell line dependent molecular profile A comparative analysis distinguished sEVs and lEVs released by temozolomide (TMZ)-treated and non TMZ-treated GBM cells on the basis of quantity, size and markers expression. Finally, the GBM-derived sEVs and lEVs, irrespective of TMZ treatment, when challenged with macrophages, modulated cell activation toward a tendentially M2b-like phenotype.
Collapse
|
20
|
Tenascin-C Function in Glioma: Immunomodulation and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:149-172. [PMID: 32845507 DOI: 10.1007/978-3-030-48457-6_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
First identified in the 1980s, tenascin-C (TNC) is a multi-domain extracellular matrix glycoprotein abundantly expressed during the development of multicellular organisms. TNC level is undetectable in most adult tissues but rapidly and transiently induced by a handful of pro-inflammatory cytokines in a variety of pathological conditions including infection, inflammation, fibrosis, and wound healing. Persistent TNC expression is associated with chronic inflammation and many malignancies, including glioma. By interacting with its receptor integrin and a myriad of other binding partners, TNC elicits context- and cell type-dependent function to regulate cell adhesion, migration, proliferation, and angiogenesis. TNC operates as an endogenous activator of toll-like receptor 4 and promotes inflammatory response by inducing the expression of multiple pro-inflammatory factors in innate immune cells such as microglia and macrophages. In addition, TNC drives macrophage differentiation and polarization predominantly towards an M1-like phenotype. In contrast, TNC shows immunosuppressive function in T cells. In glioma, TNC is expressed by tumor cells and stromal cells; high expression of TNC is correlated with tumor progression and poor prognosis. Besides promoting glioma invasion and angiogenesis, TNC has been found to affect the morphology and function of tumor-associated microglia/macrophages in glioma. Clinically, TNC can serve as a biomarker for tumor progression; and TNC antibodies have been utilized as an adjuvant agent to deliver anti-tumor drugs to target glioma. A better mechanistic understanding of how TNC impacts innate and adaptive immunity during tumorigenesis and tumor progression will open new therapeutic avenues to treat brain tumors and other malignancies.
Collapse
|
21
|
Holst CB, Christensen IJ, Skjøth-Rasmussen J, Hamerlik P, Poulsen HS, Johansen JS. Systemic Immune Modulation in Gliomas: Prognostic Value of Plasma IL-6, YKL-40, and Genetic Variation in YKL-40. Front Oncol 2020; 10:478. [PMID: 32363159 PMCID: PMC7180208 DOI: 10.3389/fonc.2020.00478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/17/2020] [Indexed: 01/10/2023] Open
Abstract
Background: Complex local and systemic immune dysfunction in glioblastoma (GBM) may affect survival. Interleukin (IL)-6 and YKL-40 are pleiotropic biomarkers present in the tumor microenvironment and involved in immune regulation. We therefore analyzed plasma IL-6, YKL-40, and genetic variation in YKL-40 and explored their ability to distinguish between glioma subtypes and predict survival in GBM. Methods: One hundred fifty-eight patients with glioma WHO grade II-IV were included in the study. Plasma collected at surgery was analyzed for IL-6 and YKL-40 (CHI3L1) by ELISA. CHI3L1 rs4950928 genotyping was analyzed on whole-blood DNA. Results: Neither plasma IL-6 nor YKL-40 corrected for age or rs4950928 genotype could differentiate GBM from lower grade gliomas. GC and GG rs4950928 genotype were associated with lower plasma YKL-40 levels (CC vs. GC, p = 0.0019; CC vs. GG, p = 0.01). Only 10 and 14 out of 94 patients with newly diagnosed GBM had elevated IL-6 or YKL-40, respectively. Most patients received corticosteroid treatment at time of blood-sampling. Higher pretreatment plasma IL-6 was associated with short overall survival (OS) [HR = 1.19 (per 2-fold change), p = 0.042] in univariate analysis. The effect disappeared in multivariate analysis. rs4950928 genotype did not associate with OS [HR = 1.30, p = 0.30]. In recurrent GBM, higher YKL-40 [HR = 2.12 (per 2-fold change), p = 0.0005] but not IL-6 [HR = 0.99 (per 2-fold change), p = 0.92] were associated with short OS in univariate analysis. Conclusion: In recurrent GBM high plasma YKL-40 may hold promise as a prognostic marker. In newly diagnosed GBM perioperative plasma IL-6, YKL-40, and genetic variation in YKL-40 did not associate with survival. Corticosteroid use may complicate interpretation of results.
Collapse
Affiliation(s)
- Camilla Bjørnbak Holst
- Department of Radiation Biology, Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Brain Tumor Biology, Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark.,Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Ib Jarle Christensen
- Department of Gastroenterology, Hvidovre Hospital, Copenhagen University Hospital, Hvidovre, Denmark
| | - Jane Skjøth-Rasmussen
- Department of Neurosurgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Petra Hamerlik
- Brain Tumor Biology, Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department of Radiation Biology, Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Julia Sidenius Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Sharabi S, Guez D, Daniels D, Cooper I, Atrakchi D, Liraz-Zaltsman S, Last D, Mardor Y. The application of point source electroporation and chemotherapy for the treatment of glioma: a randomized controlled rat study. Sci Rep 2020; 10:2178. [PMID: 32034261 PMCID: PMC7005896 DOI: 10.1038/s41598-020-59152-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/23/2020] [Indexed: 11/28/2022] Open
Abstract
The prognosis of Glioblastoma Multiforme patients is poor despite aggressive therapy. Reasons include poor chemotherapy penetration across the blood-brain barrier and tumor infiltration into surrounding tissues. Here we studied the effects of combined point-source electroporation (EP) and systemic chemotherapy in glioma-bearing rats. 128 rats were studied. Treatment groups were administered systemic Cisplatin/Methotrexate before EP (either 90 or 180 pulses). Control groups were treated by EP, chemotherapy, or no treatment. Tumor volumes were determined by MRI. Tumors growth rates of the EP + Methotrexate group (1.02 ± 0.77) were significantly lower (p < 0.01) than the control (5.2 ± 1.0) 1-week post treatment. No significant difference was found compared to Methotrexate (1.7 ± 0.5). Objective response rates (ORR) were 40% and 57% for the Methotrexate and EP + Methotrexate groups respectively. Tumor growth rates and ORR of the EP + Cisplatin groups (90 pulses 0.98 ± 0.2, 57%, 180 pulses 1.2 ± 0.1, 33%) were significantly smaller than the control (6.4 ± 1.0, p < 0.01, p < 0.02, 0%) and Cisplatin (3.9 ± 1.0, p < 0.04, p < 0.05, 13%) groups. No significant differences were found between the control groups. Increased survival was found in the EP + Cisplatin group, Χ2 = 7.54, p < 0.006 (Log Rank). Point-source EP with systemic chemotherapy is a rapid, minimal-invasive treatment that was found to induce significant antineoplastic effects in a rat glioma model.
Collapse
Affiliation(s)
- Shirley Sharabi
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel.
| | - David Guez
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel
| | - Dianne Daniels
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Interdisciplinary Center Herzliya, Herzliya, Israel
| | - Dana Atrakchi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Sigal Liraz-Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Gand Faculty of Health Profession, Ono Academic College, Kiryat Ono, Israel.,Department of Pharmacology, Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - David Last
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel
| | - Yael Mardor
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
23
|
Vidyarthi A, Agnihotri T, Khan N, Singh S, Tewari MK, Radotra BD, Chatterjee D, Agrewala JN. Predominance of M2 macrophages in gliomas leads to the suppression of local and systemic immunity. Cancer Immunol Immunother 2019; 68:1995-2004. [PMID: 31690954 DOI: 10.1007/s00262-019-02423-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 10/23/2019] [Indexed: 02/03/2023]
Abstract
Glioblastoma is a highly prevalent and aggressive form of primary brain tumor. It represents approximately 56% of all the newly diagnosed gliomas. Macrophages are one of the major constituents of tumor-infiltrating immune cells in the human gliomas. The role of immunosuppressive macrophages is very well documented in correlation with the poor prognosis of patients suffering from breast, prostate, bladder and cervical cancers. The current study highlights the correlation between the tumor-associated macrophage phenotypes and glioma progression. We observed an increase in the pool of M2 macrophages in high-grade gliomas, as confirmed by their CD68 and CD163 double-positive phenotype. In contrast, less M1 macrophages were noticed in high-grade gliomas, as evidenced by the down-regulation in the expression of CCL3 marker. In addition, we observed that higher gene expression ratio of CD163/CCL3 is associated with glioma progression. The Kaplan-Meier survival plots indicate that glioma patients with lower expression of M2c marker (CD163), and higher expression of M1 marker (CCL3) had better survival. Furthermore, we examined the systemic immune response in the peripheral blood and noted a predominance of M2 macrophages, myeloid-derived suppressor cells and PD-1+ CD4 T cells in glioma patients. Thus, the study indicates a high gene expression ratio of CD163/CCL3 in high-grade gliomas as compared to low-grade gliomas and significantly elevated frequency of M2 macrophages and PD-1+ CD4 T cells in the blood of tumor patients. These parameters could be used as an indicator of the early diagnosis and prognosis of the disease.
Collapse
Affiliation(s)
- Aurobind Vidyarthi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Department of Rheumatology, Yale University School of Medicine, New Haven, CT, USA
| | - Tapan Agnihotri
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nargis Khan
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Manoj K Tewari
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bishan D Radotra
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepyan Chatterjee
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India. .,Indian Institute of Technology, CBME Office: Room No. 115, Ropar, Punjab, 140001, India.
| |
Collapse
|
24
|
Graner MW. Roles of Extracellular Vesicles in High-Grade Gliomas: Tiny Particles with Outsized Influence. Annu Rev Genomics Hum Genet 2019; 20:331-357. [PMID: 30978305 DOI: 10.1146/annurev-genom-083118-015324] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High-grade gliomas, particularly glioblastomas (grade IV), are devastating diseases with dismal prognoses; afflicted patients seldom live longer than 15 months, and their quality of life suffers immensely. Our current standard-of-care therapy has remained essentially unchanged for almost 15 years, with little new therapeutic progress. We desperately need a better biologic understanding of these complicated tumors in a complicated organ. One area of rejuvenated study relates to extracellular vesicles (EVs)-membrane-enclosed nano- or microsized particles that originate from the endosomal system or are shed from the plasma membrane. EVs contribute to tumor heterogeneity (including the maintenance of glioma stem cells or their differentiation), the impacts of hypoxia (angiogenesis and coagulopathies), interactions amid the tumor microenvironment (concerning the survival of astrocytes, neurons, endothelial cells, blood vessels, the blood-brain barrier, and the ensuing inflammation), and influences on the immune system (both stimulatory and suppressive). This article reviews glioma EVs and the ways that EVs manifest themselves as autocrine, paracrine, and endocrine factors in proximal and distal intra- and intercellular communications. The reader should note that there is much controversy, and indeed confusion, in the field over the exact roles for EVs in many biological processes, and we will engage some of these difficulties herein.
Collapse
Affiliation(s)
- Michael W Graner
- Department of Neurosurgery, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado 80045, USA;
| |
Collapse
|
25
|
González-Morales A, Lachén-Montes M, Fernández-Irigoyen J, Santamaría E. Monitoring the Cerebrospinal Fluid Cytokine Profile Using Membrane-Based Antibody Arrays. Methods Mol Biol 2019; 2044:233-246. [PMID: 31432416 DOI: 10.1007/978-1-4939-9706-0_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The brain is the most complex organ of the human body, and the study of the different diseases and injuries that affect it is far behind the ones that affect other organs. Some of these pathologies such as neurodegenerative diseases, physical injuries, and cancer present an important alteration in its inflammatory component, which affects their outcome in a positive or negative way. For this reason, it is important to characterize the joint expression of the cytokines and growth factors (GF) that are part of this inflammatory component. The cerebrospinal fluid (CSF) is in direct contact with the brain and spinal cord, being the best biofluid to study the cytokine and GF secretion patterns of these conditions. Currently, the proteomic workflows based on mass spectrometry (MS) are unable to easily detect these proteins in CSF. In this chapter, we describe a method based on cytokine membrane arrays to characterize, in a straightforward way, the secretion profile of different cytokines and GF at once in CSF.
Collapse
Affiliation(s)
- Andrea González-Morales
- Proteomics Unit, Clinical Neuroproteomics Laboratory, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Proteored-ISCIII, Pamplona, Spain
| | - Mercedes Lachén-Montes
- Proteomics Unit, Clinical Neuroproteomics Laboratory, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Proteored-ISCIII, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Unit, Clinical Neuroproteomics Laboratory, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Proteored-ISCIII, Pamplona, Spain
| | - Enrique Santamaría
- Proteomics Unit, Clinical Neuroproteomics Laboratory, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Proteored-ISCIII, Pamplona, Spain.
| |
Collapse
|
26
|
Golán I, Rodríguez de la Fuente L, Costoya JA. NK Cell-Based Glioblastoma Immunotherapy. Cancers (Basel) 2018; 10:E522. [PMID: 30567306 PMCID: PMC6315402 DOI: 10.3390/cancers10120522] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/01/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GB) is the most aggressive and most common malignant primary brain tumor diagnosed in adults. GB shows a poor prognosis and, unfortunately, current therapies are unable to improve its clinical outcome, imposing the need for innovative therapeutic approaches. The main reason for the poor prognosis is the great cell heterogeneity of the tumor mass and its high capacity for invading healthy tissues. Moreover, the glioblastoma microenvironment is capable of suppressing the action of the immune system through several mechanisms such as recruitment of cell modulators. Development of new therapies that avoid this immune evasion could improve the response to the current treatments for this pathology. Natural Killer (NK) cells are cellular components of the immune system more difficult to deceive by tumor cells and with greater cytotoxic activity. Their use in immunotherapy gains strength because they are a less toxic alternative to existing therapy, but the current research focuses on mimicking the NK attack strategy. Here, we summarize the most recent studies regarding molecular mechanisms involved in the GB and immune cells interaction and highlight the relevance of NK cells in the new therapeutic challenges.
Collapse
Affiliation(s)
- Irene Golán
- Molecular Oncology Laboratory MOL, Departamento de Fisioloxia, CiMUS, Facultade de Medicina, Universidade de Santiago de Compostela, IDIS, 15782 Santiago de Compostela, Spain.
| | - Laura Rodríguez de la Fuente
- Molecular Oncology Laboratory MOL, Departamento de Fisioloxia, CiMUS, Facultade de Medicina, Universidade de Santiago de Compostela, IDIS, 15782 Santiago de Compostela, Spain.
| | - Jose A Costoya
- Molecular Oncology Laboratory MOL, Departamento de Fisioloxia, CiMUS, Facultade de Medicina, Universidade de Santiago de Compostela, IDIS, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
27
|
Histological and Immunohistological Features of Reccurences in Patients with High Grade Diffuse Astrocytic Tumors. Fam Med 2018. [DOI: 10.30841/2307-5112.4.2018.163293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Rapp M, Grauer OM, Kamp M, Sevens N, Zotz N, Sabel M, Sorg RV. A randomized controlled phase II trial of vaccination with lysate-loaded, mature dendritic cells integrated into standard radiochemotherapy of newly diagnosed glioblastoma (GlioVax): study protocol for a randomized controlled trial. Trials 2018; 19:293. [PMID: 29801515 PMCID: PMC5970474 DOI: 10.1186/s13063-018-2659-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/02/2018] [Indexed: 01/06/2023] Open
Abstract
Background Despite the combination of surgical resection, radio- and chemotherapy, median survival of glioblastoma multiforme (GBM) patients only slightly increased in the last years. Disease recurrence is definite with no effective therapy existing after tumor removal. Dendritic cell (DC) vaccination is a promising active immunotherapeutic approach. There is clear evidence that it is feasible, results in immunological anti-tumoral responses, and appears to be beneficial for survival and quality of life of GBM patients. Moreover, combining it with the standard therapy of GBM may allow exploiting synergies between the treatment modalities. In this randomized controlled trial, we seek to confirm these promising initial results. Methods One hundred and thirty-six newly diagnosed, isocitrate dehydrogenase wildtype GBM patients will be randomly allocated (1:1 ratio, stratified by O6-methylguanine-DNA-methyltransferase promotor methylation status) after near-complete resection in a multicenter, prospective phase II trial into two groups: (1) patients receiving the current therapeutic “gold standard” of radio/temozolomide chemotherapy and (2) patients receiving DC vaccination as an add-on to the standard therapy. A recruitment period of 30 months is anticipated; follow-up will be 2 years. The primary objective of the study is to compare overall survival (OS) between the two groups. Secondary objectives are comparing progression-free survival (PFS) and 6-, 12- and 24-month OS and PFS rates, the safety profile, overall and neurological performance and quality of life. Discussion Until now, close to 500 GBM patients have been treated with DC vaccination in clinical trials or on a compassionate-use basis. Results have been encouraging, but cannot provide robust evidence of clinical efficacy because studies have been non-controlled or patient numbers have been low. Therefore, a prospective, randomized phase II trial with a sufficiently large number of patients is now mandatory for clear evidence regarding the impact of DC vaccination on PFS and OS in GBM. Trial registration Protocol code: GlioVax, date of registration: 17. February 2017. Trial identifier: EudraCT-Number 2017–000304-14. German Registry for Clinical Studies, ID: DRKS00013248 (approved primary register in the WHO network) and at ClinicalTrials.gov, ID: NCT03395587. Registered on 11 March 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2659-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marion Rapp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Department of Neurosurgery, Heinrich Heine University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - Oliver M Grauer
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Marcel Kamp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Natalie Sevens
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Nikola Zotz
- Coordination Center for Clinical Trials, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Michael Sabel
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| |
Collapse
|
29
|
Quezada C, Torres Á, Niechi I, Uribe D, Contreras-Duarte S, Toledo F, San Martín R, Gutiérrez J, Sobrevia L. Role of extracellular vesicles in glioma progression. Mol Aspects Med 2018; 60:38-51. [PMID: 29222067 DOI: 10.1016/j.mam.2017.12.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022]
Abstract
The role of extracellular vesicles in cancer biology has emerged as a focus of the study of great importance and has been shown to directly influence tumour development in several cancers including brain tumours, such as gliomas. Gliomas are the most aggressive brain tumours, and in the last time, a considerable effort has been made to understand their biology. Studies focus in the signalling pathways involved in the processes of angiogenesis, viability, drug resistance and immune response evasion, as well as gliomas ability to infiltrate healthy tissue, a phenomenon regulated by the migratory and invasive capacity of the cells within a tumour. In this review, we summarize the different types and classifications of extracellular vesicles, their intravesicular content, and their role in the regulation of tumour progression processes in glioma.
Collapse
Affiliation(s)
- Claudia Quezada
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile.
| | - Ángelo Torres
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Niechi
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Uribe
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Susana Contreras-Duarte
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile
| | - Rody San Martín
- Molecular Pathology Laboratory, Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Jaime Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastián, Santiago 7510157, Chile.
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia.
| |
Collapse
|
30
|
Guadagno E, Presta I, Maisano D, Donato A, Pirrone CK, Cardillo G, Corrado SD, Mignogna C, Mancuso T, Donato G, Del Basso De Caro M, Malara N. Role of Macrophages in Brain Tumor Growth and Progression. Int J Mol Sci 2018; 19:ijms19041005. [PMID: 29584702 PMCID: PMC5979398 DOI: 10.3390/ijms19041005] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 12/16/2022] Open
Abstract
The role of macrophages in the growth and the progression of tumors has been extensively studied in recent years. A large body of data demonstrates that macrophage polarization plays an essential role in the growth and progression of brain tumors, such as gliomas, meningiomas, and medulloblastomas. The brain neoplasm cells have the ability to influence the polarization state of the tumor associated macrophages. In turn, innate immunity cells have a decisive role through regulation of the acquired immune response, but also through humoral cross-talking with cancer cells in the tumor microenvironment. Neoangiogenesis, which is an essential element in glial tumor progression, is even regulated by the tumor associated macrophages, whose activity is linked to other factors, such as hypoxia. In addition, macrophages play a decisive role in establishing the entry into the bloodstream of cancer cells. As is well known, the latter phenomenon is also present in brain tumors, even if they only rarely metastasize. Looking ahead in the future, we can imagine that characterizing the relationships between tumor and tumor associated macrophage, as well as the study of circulating tumor cells, could give us useful tools in prognostic evaluation and therapy. More generally, the study of innate immunity in brain tumors can boost the development of new forms of immunotherapy.
Collapse
Affiliation(s)
- Elia Guadagno
- Department of Advanced Biomedical Sciences-Pathology Section, University of Naples "Federico II"-via Pansini 5, 80131 Naples, Italy.
| | - Ivan Presta
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Domenico Maisano
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Annalidia Donato
- Department of Medical and Surgical Sciences-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| | - Caterina Krizia Pirrone
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Gabriella Cardillo
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Simona Domenica Corrado
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Chiara Mignogna
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Teresa Mancuso
- Department of Medical and Surgical Sciences-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| | - Giuseppe Donato
- Department of Health Sciences, University of Catanzaro "Magna Græcia"-viale Europa, 88100 Catanzaro, Italy.
| | - Marialaura Del Basso De Caro
- Department of Advanced Biomedical Sciences-Pathology Section, University of Naples "Federico II"-via Pansini 5, 80131 Naples, Italy.
| | - Natalia Malara
- Department of Clinical and Experimental Medicine-University of Catanzaro "Magna Graecia"-viale Europa, 88100 Catanzaro, Italy.
| |
Collapse
|
31
|
Chistiakov DA, Chekhonin IV, Chekhonin VP. The EGFR variant III mutant as a target for immunotherapy of glioblastoma multiforme. Eur J Pharmacol 2017; 810:70-82. [DOI: 10.1016/j.ejphar.2017.05.064] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/15/2017] [Accepted: 05/31/2017] [Indexed: 12/26/2022]
|
32
|
Cheng Z, Zhang D, Gong B, Wang P, Liu F. CD163 as a novel target gene of STAT3 is a potential therapeutic target for gastric cancer. Oncotarget 2017; 8:87244-87262. [PMID: 29152078 PMCID: PMC5675630 DOI: 10.18632/oncotarget.20244] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/18/2017] [Indexed: 12/15/2022] Open
Abstract
CD163 is a member of the scavenger receptor cysteine-rich superfamily, and has been widely used to identify M2 type macrophage. However, the expression of CD163 in gastric cancer and its regulatory mechanism are still unclear. Here we show that CD163 is elevated in gastric cancer tissues. High expression of CD163 is a potential indicator to evaluate the status of tumor associated macrophages (TAMs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs) and cancer associated fibroblasts (Cafs). Besides, more CD163 positive macrophages and CD163 expressing gastric cancer cells are associated with tumor invasion and poor prognosis. Knocking-down CD163 in cancer cells could inhibit tumor growth in vivo. We also find various immune molecules which are correlated with CD163 in gastric cancer tissues and cell lines have positive staining in the cancer cells of clinical sample. Finally, we confirm CD163 is a novel target gene of STAT3 (signal transducer and activator of transcription 3) in gastric cancer. Our data indicate that CD163 may be a potential poor prognostic marker and therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Zhenguo Cheng
- National Center for The International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Danhua Zhang
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Baocheng Gong
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Pengliang Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Funan Liu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
33
|
Chitadze G, Flüh C, Quabius ES, Freitag-Wolf S, Peters C, Lettau M, Bhat J, Wesch D, Oberg HH, Luecke S, Janssen O, Synowitz M, Held-Feindt J, Kabelitz D. In-depth immunophenotyping of patients with glioblastoma multiforme: Impact of steroid treatment. Oncoimmunology 2017; 6:e1358839. [PMID: 29147621 DOI: 10.1080/2162402x.2017.1358839] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/16/2017] [Accepted: 07/19/2017] [Indexed: 01/01/2023] Open
Abstract
Despite aggressive treatment regimens based on surgery and radiochemotherapy, the prognosis of patients with grade IV glioblastoma multiforme (GBM) remains extremely poor, calling for alternative options such as immunotherapy. Immunological mechanisms including the Natural Killer Group 2 member D (NKG2D) receptor-ligand system play an important role in tumor immune surveillance and targeting the NKG2D system might be beneficial. However, before considering any kind of immunotherapy, a precise characterization of the immune system is important, particularly in GBM patients where conventional therapies with impact on the immune system are frequently co-administered. Here we performed an in-depth immunophenotyping of GBM patients and age-matched healthy controls and analyzed NKG2D ligand expression on primary GBM cells ex vivo. We report that GBM patients have a compromised innate immune system irrespective of steroid (dexamethasone) medication. However, dexamethasone drastically reduced the number of immune cells in the blood of GBM patients. Moreover, higher counts of immune cells influenced by dexamethasone like CD45+ lymphocytes and non-Vδ2 γδ T cells were associated with better overall survival. Higher levels of NKG2D ligands on primary GBM tumor cells were observed in patients who received radiochemotherapy, pointing towards increased immunogenic potential of GBM cells following standard radiochemotherapy. This study sheds light on how steroids and radiochemotherapy affect immune cell parameters of GBM patients, a pre-requisite for the development of new therapeutic strategies targeting the immune system in these patients.
Collapse
Affiliation(s)
- Guranda Chitadze
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Charlotte Flüh
- Dept. of Neurosurgery, UKSH Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Elgar Susanne Quabius
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany.,Dept. of Oto-Rhino-Laryngology, UKSH Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, Kiel University, Kiel, Schleswig-Hostein, Germany
| | - Christian Peters
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Marcus Lettau
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Jaydeep Bhat
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Daniela Wesch
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Hans-Heinrich Oberg
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Stefanie Luecke
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Ottmar Janssen
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Michael Synowitz
- Dept. of Neurosurgery, UKSH Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Janka Held-Feindt
- Dept. of Neurosurgery, UKSH Campus Kiel, Kiel, Schleswig-Hostein, Germany
| | - Dieter Kabelitz
- Institute of Immunology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Schleswig-Hostein, Germany
| |
Collapse
|
34
|
Schiffer D, Mellai M, Bovio E, Annovazzi L. The neuropathological basis to the functional role of microglia/macrophages in gliomas. Neurol Sci 2017; 38:1571-1577. [PMID: 28593528 DOI: 10.1007/s10072-017-3002-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022]
Abstract
The paper wants to be a tracking shot of the main recent acquisitions on the function and significance of microglia/macrophages in gliomas. The observations have been principally carried out on in vitro cultures and on tumor transplants in animals. Contrary to what is deduced from microglia in non-neoplastic pathologic conditions of central nervous system (CNS), most conclusions indicate that microglia acts favoring tumor proliferation through an immunosuppression induced by glioma cells. By immunohistochemistry, different microglia phenotypes are recognized in gliomas, from ramified microglia to frank macrophagic aspect. One wonders whether the functional conclusions drawn from many microglia studies, but not in conditions of human pathology, apply to all the phenotypes recognizable in them. It is difficult to verify in human pathology a prognostic significance of microglia. Only CD163-positive microglia/macrophages inversely correlate with glioma patients' survival, whereas the total number of microglia does not change with the malignancy grade.
Collapse
Affiliation(s)
- Davide Schiffer
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy.
| | - Marta Mellai
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| | - Enrica Bovio
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| | - Laura Annovazzi
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| |
Collapse
|
35
|
Liu D, Tian S, Zhang K, Xiong W, Lubaki NM, Chen Z, Han W. Chimeric antigen receptor (CAR)-modified natural killer cell-based immunotherapy and immunological synapse formation in cancer and HIV. Protein Cell 2017; 8:861-877. [PMID: 28488245 PMCID: PMC5712291 DOI: 10.1007/s13238-017-0415-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells contribute to the body’s immune defenses. Current chimeric antigen receptor (CAR)-modified T cell immunotherapy shows strong promise for treating various cancers and infectious diseases. Although CAR-modified NK cell immunotherapy is rapidly gaining attention, its clinical applications are mainly focused on preclinical investigations using the NK92 cell line. Despite recent advances in CAR-modified T cell immunotherapy, cost and severe toxicity have hindered its widespread use. To alleviate these disadvantages of CAR-modified T cell immunotherapy, additional cytotoxic cell-mediated immunotherapies are urgently needed. The unique biology of NK cells allows them to serve as a safe, effective, alternative immunotherapeutic strategy to CAR-modified T cells in the clinic. While the fundamental mechanisms underlying the cytotoxicity and side effects of CAR-modified T and NK cell immunotherapies remain poorly understood, the formation of the immunological synapse (IS) between CAR-modified T or NK cells and their susceptible target cells is known to be essential. The role of the IS in CAR T and NK cell immunotherapies will allow scientists to harness the power of CAR-modified T and NK cells to treat cancer and infectious diseases. In this review, we highlight the potential applications of CAR-modified NK cells to treat cancer and human immunodeficiency virus (HIV), and discuss the challenges and possible future directions of CAR-modified NK cell immunotherapy, as well as the importance of understanding the molecular mechanisms of CAR-modified T cell- or NK cell-mediated cytotoxicity and side effects, with a focus on the CAR-modified NK cell IS.
Collapse
Affiliation(s)
- Dongfang Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| | - Shuo Tian
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kai Zhang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Wei Xiong
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ndongala Michel Lubaki
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Zhiying Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Weidong Han
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
36
|
Hao W, Luo W, Bai M, Li J, Bai X, Guo J, Wu J, Wang M. MicroRNA-206 Inhibited the Progression of Glioblastoma Through BCL-2. J Mol Neurosci 2016; 60:531-538. [PMID: 27558109 DOI: 10.1007/s12031-016-0824-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 08/17/2016] [Indexed: 12/21/2022]
Abstract
Gliomas are the most common type of brain tumor and have a poor prognosis. MicroRNAs (miRNAs) are a class of small, endogenous, and non-coding RNAs that play crucial roles in cell proliferation, survival, and invasion. Deregulated expression of miR-206 has been investigated in many cancers. However, the role of miR-206 in glioblastoma is still unclear. In the present study, we found that the expression of miR-206 was decreased in cancer tissues compared with normal tissues. However, the expression level of BCL-2 was higher in cancer tissues than that in normal tissues (all p < 0.001). Statistically, the expression level of BCL-2 was inversely correlated with the miR-206. In addition, the overall survival of glioblastoma patients with lower miR-206 expression was significantly shorter than those with high miR-206 expression (p < 0.001). Besides, the expression of miR-206 was also decreased in U87 and U251 cells. In vitro assays showed that ectopic miR-206 expression affected the proliferation, cell cycle, and invasion in U87 and U251 cells. Importantly, we identified BCL-2 as a direct target of miR-206 in U87 and U251 cells using luciferase assay. Overexpression of BCL-2 partially attenuated the miR-206-mediated cell proliferation. In vivo, overexpression of miR-206 suppressed the progression of glioblastoma cells using mice xenograft model. In conclusion, this study suggested that miR-206 could act as a tumor suppressor gene through inhibiting BCL-2 in the development of glioblastoma.
Collapse
Affiliation(s)
- Wenjiong Hao
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.,Department of Neurosurgery, Affiliated Hospital, Medical College of Yan'an University, Yan'an City, Shaanxi, 716000, People's Republic of China
| | - Wei Luo
- Department of Neurosurgery, The Affiliated Hospital of Shaanxi Traditional Chinese Medicine University, Xianyang, 712000, People's Republic of China
| | - Mangmang Bai
- Department of Neurosurgery, Affiliated Hospital, Medical College of Yan'an University, Yan'an City, Shaanxi, 716000, People's Republic of China
| | - Jian Li
- Department of Neurosurgery, Affiliated Hospital, Medical College of Yan'an University, Yan'an City, Shaanxi, 716000, People's Republic of China
| | - Xiaobin Bai
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jie Guo
- Department of Neurosurgery, Affiliated Hospital, Medical College of Yan'an University, Yan'an City, Shaanxi, 716000, People's Republic of China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| |
Collapse
|