1
|
Lu J, Huo W, Ma Y, Wang X, Yu J. Suppressive immune microenvironment and CART therapy for glioblastoma: Future prospects and challenges. Cancer Lett 2024; 600:217185. [PMID: 39142498 DOI: 10.1016/j.canlet.2024.217185] [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] [Received: 05/10/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Glioblastoma, a highly malignant intracranial tumor, has acquired slow progress in treatment. Previous clinical trials involving targeted therapy and immune checkpoint inhibitors have shown no significant benefits in treating glioblastoma. This ineffectiveness is largely due to the complex immunosuppressive environment of glioblastoma. Glioblastoma cells exhibit low immunogenicity and strong heterogeneity and the immune microenvironment is replete with inhibitory cytokines, numerous immunosuppressive cells, and insufficient effective T cells. Fortunately, recent Phase I clinical trials of CART therapy for glioblastoma have confirmed its safety, with a small subset of patients achieving survival benefits. However, CART therapy continues to face challenges, including blood-brain barrier obstruction, antigen loss, and an immunosuppressive tumor microenvironment (TME). This article provides a detailed examination of glioblastoma's immune microenvironment, both from intrinsic and extrinsic tumor cell factors, reviews current clinical and basic research on multi-targets CART treatment, and concludes by outlining the key challenges in using CART cells for glioblastoma therapy.
Collapse
Affiliation(s)
- Jie Lu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Wen Huo
- Department of Radiation Oncology, Affiliated Tumor Hospital of Xinjiang Medical University, China
| | - Yingze Ma
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, Shandong, China; Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong, China
| | - Xin Wang
- Department of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
2
|
Guo H, Xu X, Zhang J, Du Y, Yang X, He Z, Zhao L, Liang T, Guo L. The Pivotal Role of Preclinical Animal Models in Anti-Cancer Drug Discovery and Personalized Cancer Therapy Strategies. Pharmaceuticals (Basel) 2024; 17:1048. [PMID: 39204153 PMCID: PMC11357454 DOI: 10.3390/ph17081048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
The establishment and utilization of preclinical animal models constitute a pivotal aspect across all facets of cancer research, indispensably contributing to the comprehension of disease initiation and progression mechanisms, as well as facilitating the development of innovative anti-cancer therapeutic approaches. These models have emerged as crucial bridges between basic and clinical research, offering multifaceted support to clinical investigations. This study initially focuses on the importance and benefits of establishing preclinical animal models, discussing the different types of preclinical animal models and recent advancements in cancer research. It then delves into cancer treatment, studying the characteristics of different stages of tumor development and the development of anti-cancer drugs. By integrating tumor hallmarks and preclinical research, we elaborate on the path of anti-cancer drug development and provide guidance on personalized cancer therapy strategies, including synthetic lethality approaches and novel drugs widely adopted in the field. Ultimately, we summarize a strategic framework for selecting preclinical safety experiments, tailored to experimental modalities and preclinical animal species, and present an outlook on the prospects and challenges associated with preclinical animal models. These models undoubtedly offer new avenues for cancer research, encompassing drug development and personalized anti-cancer protocols. Nevertheless, the road ahead continues to be lengthy and fraught with obstacles. Hence, we encourage researchers to persist in harnessing advanced technologies to refine preclinical animal models, thereby empowering these emerging paradigms to positively impact cancer patient outcomes.
Collapse
Affiliation(s)
- Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Xinru Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Yajing Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Xinbing Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Zhiheng He
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (Z.H.); (L.Z.)
| | - Linjie Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (Z.H.); (L.Z.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (Z.H.); (L.Z.)
| |
Collapse
|
3
|
Pesini C, Artal L, Paúl Bernal J, Sánchez Martinez D, Pardo J, Ramírez-Labrada A. In-depth analysis of the interplay between oncogenic mutations and NK cell-mediated cancer surveillance in solid tumors. Oncoimmunology 2024; 13:2379062. [PMID: 39036370 PMCID: PMC11259085 DOI: 10.1080/2162402x.2024.2379062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Natural killer (NK) cells play a crucial role in antitumoral and antiviral responses. Yet, cancer cells can alter themselves or the microenvironment through the secretion of cytokines or other factors, hindering NK cell activation and promoting a less cytotoxic phenotype. These resistance mechanisms, often referred to as the "hallmarks of cancer" are significantly influenced by the activation of oncogenes, impacting most, if not all, of the described hallmarks. Along with oncogenes, other types of genes, the tumor suppressor genes are frequently mutated or modified during cancer. Traditionally, these genes have been associated with uncontrollable tumor growth and apoptosis resistance. Recent evidence suggests oncogenic mutations extend beyond modulating cell death/proliferation programs, influencing cancer immunosurveillance. While T cells have been more studied, the results obtained highlight NK cells as emerging key protagonists for enhancing tumor cell elimination by modulating oncogenic activity. A few recent studies highlight the crucial role of oncogenic mutations in NK cell-mediated cancer recognition, impacting angiogenesis, stress ligands, and signaling balance within the tumor microenvironment. This review will critically examine recent discoveries correlating oncogenic mutations to NK cell-mediated cancer immunosurveillance, a relatively underexplored area, particularly in the era dominated by immune checkpoint inhibitors and CAR-T cells. Building on these insights, we will explore opportunities to improve NK cell-based immunotherapies, which are increasingly recognized as promising alternatives for treating low-antigenic tumors, offering significant advantages in terms of safety and manufacturing suitability.
Collapse
Affiliation(s)
- Cecilia Pesini
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Laura Artal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
| | - Jorge Paúl Bernal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Diego Sánchez Martinez
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ariel Ramírez-Labrada
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
| |
Collapse
|
4
|
Pérez de la Cruz G, Pérez de la Cruz V, Navarro Cossio J, Vázquez Cervantes GI, Salazar A, Orozco Morales M, Pineda B. Kynureninase Promotes Immunosuppression and Predicts Survival in Glioma Patients: In Silico Data Analyses of the Chinese Glioma Genome Atlas (CGGA) and of the Cancer Genome Atlas (TCGA). Pharmaceuticals (Basel) 2023; 16:ph16030369. [PMID: 36986469 PMCID: PMC10051585 DOI: 10.3390/ph16030369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Kynureninase (KYNU) is a kynurenine pathway (KP) enzyme that produces metabolites with immunomodulatory properties. In recent years, overactivation of KP has been associated with poor prognosis of several types of cancer, in particular by promoting the invasion, metastasis, and chemoresistance of cancer cells. However, the role of KYNU in gliomas remains to be explored. In this study, we used the available data from TCGA, CGGA and GTEx projects to analyze KYNU expression in gliomas and healthy tissue, as well as the potential contribution of KYNU in the tumor immune infiltrate. In addition, immune-related genes were screened with KYNU expression. KYNU expression correlated with the increased malignancy of astrocytic tumors. Survival analysis in primary astrocytomas showed that KYNU expression correlated with poor prognosis. Additionally, KYNU expression correlated positively with several genes related to an immunosuppressive microenvironment and with the characteristic immune tumor infiltrate. These findings indicate that KYNU could be a potential therapeutic target for modulating the tumor microenvironment and enhancing an effective antitumor immune response.
Collapse
Affiliation(s)
- Gonzalo Pérez de la Cruz
- Department of Mathematics, Faculty of Sciences, Universidad Nacional Autónoma de México, UNAM, Mexico City 04510, Mexico
| | - Verónica Pérez de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Javier Navarro Cossio
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Gustavo Ignacio Vázquez Cervantes
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Aleli Salazar
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Mario Orozco Morales
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Benjamin Pineda
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
- Correspondence: ; Tel.: +52-55-5606-4040
| |
Collapse
|
5
|
Natural killer cell awakening: unleash cancer-immunity cycle against glioblastoma. Cell Death Dis 2022; 13:588. [PMID: 35803912 PMCID: PMC9270460 DOI: 10.1038/s41419-022-05041-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/25/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Due to the negligence of the complex tumor immune microenvironment, traditional treatment for glioblastoma has reached its limitation and cannot achieve a satisfying outcome in the past decade. The emergence of immunotherapy based on the theory of cancer-immunity cycle has brought a new dawn to glioblastoma patients. However, the results of most phase II and phase III clinical trials are not optimistic due to the simple focus on T cells activation rather than other immune cells involved in anti-tumor immunity. NK cells play a critical role in both innate and adaptive immunity, having the ability to coordinate immune response in inflammation, autoimmune disease and cancer. They are expected to cooperate with T cells to maximize the anti-tumor immune effect and have great potential in treating glioblastoma. Here, we describe the traditional treatment methods and current immunotherapy strategies for glioblastoma. Then, we list a microenvironment map and discuss the reasons for glioblastoma inhibitory immunity from multiple perspectives. More importantly, we focus on the advantages of NK cells as potential immune regulatory cells and the ways to maximize their anti-tumor immune effect. Finally, our outlook on the directions and potential applications of NK cell-based therapy combining with the advance technologies is presented. This review depicts NK cell awakening as the precondition to unleash the cancer-immunity cycle against glioblastoma and elaborate this idea from biology to clinical treatment.
Collapse
|
6
|
Shafi O, Siddiqui G. Tracing the origins of glioblastoma by investigating the role of gliogenic and related neurogenic genes/signaling pathways in GBM development: a systematic review. World J Surg Oncol 2022; 20:146. [PMID: 35538578 PMCID: PMC9087910 DOI: 10.1186/s12957-022-02602-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/15/2022] [Indexed: 02/16/2023] Open
Abstract
Background Glioblastoma is one of the most aggressive tumors. The etiology and the factors determining its onset are not yet entirely known. This study investigates the origins of GBM, and for this purpose, it focuses primarily on developmental gliogenic processes. It also focuses on the impact of the related neurogenic developmental processes in glioblastoma oncogenesis. It also addresses why glial cells are at more risk of tumor development compared to neurons. Methods Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving glioblastoma, gliogenesis, neurogenesis, stemness, neural stem cells, gliogenic signaling and pathways, neurogenic signaling and pathways, and astrocytogenic genes. Results The origin of GBM is dependent on dysregulation in multiple genes and pathways that accumulatively converge the cells towards oncogenesis. There are multiple layers of steps in glioblastoma oncogenesis including the failure of cell fate-specific genes to keep the cells differentiated in their specific cell types such as p300, BMP, HOPX, and NRSF/REST. There are genes and signaling pathways that are involved in differentiation and also contribute to GBM such as FGFR3, JAK-STAT, and hey1. The genes that contribute to differentiation processes but also contribute to stemness in GBM include notch, Sox9, Sox4, c-myc gene overrides p300, and then GFAP, leading to upregulation of nestin, SHH, NF-κB, and others. GBM mutations pathologically impact the cell circuitry such as the interaction between Sox2 and JAK-STAT pathway, resulting in GBM development and progression. Conclusion Glioblastoma originates when the gene expression of key gliogenic genes and signaling pathways become dysregulated. This study identifies key gliogenic genes having the ability to control oncogenesis in glioblastoma cells, including p300, BMP, PAX6, HOPX, NRSF/REST, LIF, and TGF beta. It also identifies key neurogenic genes having the ability to control oncogenesis including PAX6, neurogenins including Ngn1, NeuroD1, NeuroD4, Numb, NKX6-1 Ebf, Myt1, and ASCL1. This study also postulates how aging contributes to the onset of glioblastoma by dysregulating the gene expression of NF-κB, REST/NRSF, ERK, AKT, EGFR, and others.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
7
|
Filippone A, Lanza M, Mannino D, Raciti G, Colarossi C, Sciacca D, Cuzzocrea S, Paterniti I. PD1/PD-L1 immune checkpoint as a potential target for preventing brain tumor progression. Cancer Immunol Immunother 2022; 71:2067-2075. [PMID: 35092481 PMCID: PMC9374620 DOI: 10.1007/s00262-021-03130-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023]
Abstract
Programmed death-1 (PD-1) is a cell surface receptor that functions as a T cell checkpoint and plays a central role in regulating T cell collapse. The binding of PD-1 to its ligand programmed death-ligand 1 (PD-L1) activates downstream signaling pathways and inhibits T cell activation in the perspective of immune system mechanism and regulation in tumor progression. It is well reported that tumors adopt certain immune-checkpoint pathways as a mechanism of resistance against immune cells such as T cells that are specific for tumor antigens. Indeed, the PD-1/PD-L1 pathway controls the induction and maintenance of immune tolerance within the tumor microenvironment. Thus, the PD-1/PD-L1 checkpoint regulation appears to be of extreme importance as well as the immunotherapy targeting that via and the using of PD-1/PD-L1 inhibitors that have changed the scenario of brain cancer treatment and survival. Here, we review the mechanism of action of PD-1 and PD-L1, the PD/PDL-1 signaling pathway involved in the progression of brain tumors, and its application as cancer immunotherapy counteracting tumor escape in central nervous system.
Collapse
Affiliation(s)
- A Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - M Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - D Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - G Raciti
- IOM Ricerca Srl, via Penninazzo 11, 95029, Catania, Italy
| | - C Colarossi
- Instituto Oncologico del Mediterraneo, via Penninazzo 7, 95029, Catania, Italy
| | - D Sciacca
- Instituto Oncologico del Mediterraneo, via Penninazzo 7, 95029, Catania, Italy
| | - S Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - I Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy.
| |
Collapse
|
8
|
Targeting Glioblastoma via Selective Alteration of Mitochondrial Redox State. Cancers (Basel) 2022; 14:cancers14030485. [PMID: 35158753 PMCID: PMC8833725 DOI: 10.3390/cancers14030485] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Glioblastoma is characterized by a pronounced redox imbalance due to elevated glycolytic and mitochondrial oxidative metabolism. New therapeutic strategies have been developed to modulate glioblastoma redox signaling to effectively suppress growth and prolong survival. However, drug selectivity and therapeutic relapse prove to be the major challenges. We describe a pharmacological strategy for the selective targeting and treatment of glioblastoma using the redox active combination drug menadione/ascorbate, which is characterized by tolerance to normal cells and tissues. Menadione/ascorbate treatment of glioblastoma mice suppressed tumor growth and significantly increased survival without adverse side effects. This is accompanied by increased oxidative stress, decreased reducing capacity and decreased cellular density in the tumor alone, as well as increased brain perfusion and decreased regulation of several oncoproteins and oncometabolites, which implies modulation of the immune response and reduced drug resistance. We believe that this therapeutic strategy is feasible and promising and deserves the attention of clinicians. Abstract Glioblastoma is one of the most aggressive brain tumors, characterized by a pronounced redox imbalance, expressed in a high oxidative capacity of cancer cells due to their elevated glycolytic and mitochondrial oxidative metabolism. The assessment and modulation of the redox state of glioblastoma are crucial factors that can provide highly specific targeting and treatment. Our study describes a pharmacological strategy for targeting glioblastoma using a redox-active combination drug. The experiments were conducted in vivo on glioblastoma mice (intracranial model) and in vitro on cell lines (cancer and normal) treated with the redox cycling pair menadione/ascorbate (M/A). The following parameters were analyzed in vivo using MRI or ex vivo on tissue and blood specimens: tumor growth, survival, cerebral perfusion, cellular density, tissue redox state, expression of tumor-associated NADH oxidase (tNOX) and transforming growth factor-beta 1 (TGF-β1). Dose-dependent effects of M/A on cell viability, mitochondrial functionality, and redox homeostasis were evaluated in vitro. M/A treatment suppressed tumor growth and significantly increased survival without adverse side effects. This was accompanied by increased oxidative stress, decreased reducing capacity, and decreased cellular density in the tumor only, as well as increased cerebral perfusion and down-regulation of tNOX and TGF-β1. M/A induced selective cytotoxicity and overproduction of mitochondrial superoxide in isolated glioblastoma cells, but not in normal microglial cells. This was accompanied by a significant decrease in the over-reduced state of cancer cells and impairment of their “pro-oncogenic” functionality, assessed by dose-dependent decreases in: NADH, NAD+, succinate, glutathione, cellular reducing capacity, mitochondrial potential, steady-state ATP, and tNOX expression. The safety of M/A on normal cells was compromised by treatment with cerivastatin, a non-specific prenyltransferase inhibitor. In conclusion, M/A differentiates glioblastoma cells and tissues from normal cells and tissues by redox targeting, causing severe oxidative stress only in the tumor. The mechanism is complex and most likely involves prenylation of menadione in normal cells, but not in cancer cells, modulation of the immune response, a decrease in drug resistance, and a potential role in sensitizing glioblastoma to conventional chemotherapy.
Collapse
|
9
|
Li X, Liu M, Zhao J, Ren T, Yan X, Zhang L, Wang X. Research Progress About Glioma Stem Cells in the Immune Microenvironment of Glioma. Front Pharmacol 2021; 12:750857. [PMID: 34630121 PMCID: PMC8495125 DOI: 10.3389/fphar.2021.750857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Gliomas are the most common primary tumors of the central nervous system. Due to the existence of the blood-brain barrier and its unique regional immune characteristics, the study of the immune microenvironment of gliomas is particularly important. Glioma stem cells are an important cause of initiating glioma, promoting tumor progression and leading to tumor recurrence. Immunotherapeutic strategies targeting glioma stem cells have become the focus of current research. This paper will focus on the research progress of glioma stem cells in the immune microenvironment of glioma to provide the basis for the immunotherapy of glioma.
Collapse
Affiliation(s)
- Xiangyu Li
- Department of Neurosurgery, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ming Liu
- Department of Neurosurgery, Ningde Municipal Hospital Affiliated of Ningde Normal College, Ningde, China
| | - Junfeng Zhao
- Department of Neurosurgery, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tong Ren
- Department of Neurosurgery, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Yan
- Department of Medical Oncology, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lijun Zhang
- Department of Ophthalmology, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xun Wang
- Department of Neurosurgery, The Third People’s Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Shao G, Liu Q, Yang L, Feng G, Zhao W, Huang Z, Yang Z. Prediction and identification of novel HLA-A*0201-restricted cytotoxic T lymphocyte epitopes from endocan. JOURNAL OF INFLAMMATION-LONDON 2020; 17:10. [PMID: 32099535 PMCID: PMC7031931 DOI: 10.1186/s12950-020-00240-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/11/2020] [Indexed: 11/10/2022]
Abstract
Background Prediction and identification of cytotoxic T lymphocyte (CTL) epitopes from tumor associated antigens is a crucial step for the development of tumor immunotherapy strategy. Endocan has been identified as antigen overexpressed in various tumors. Methods In this experiment, we predicted and identified HLA-A2-restricted CTL epitopes from endocan by using the following procedures. Firstly, we predicted the epitopes from the amino acid sequence of endocan by computer-based methods; Secondly, we determined the affinity of the predicted peptide with HLA-A2.1 molecule by peptide-binding assay; Thirdly, we elicited the primary T cell response against the predicted peptides in vitro; Lastly, we tested the specific CTLs toward endocan and HLA-A2.1 positive target cells. Results These data demonstrated that peptides of endocan containing residues 4–12 and 9–17 could elicit specific CTLs producing interferon-γ and cytotoxicity. Conclusions Therefore, our findings suggested that the predicted peptides were novel HLA-A2.1-restricted CTL epitopes, and might provide promising target for tumor immunotherapy.
Collapse
Affiliation(s)
- Gaohai Shao
- 1Department of orthopedics, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Qingjun Liu
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Ling Yang
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Guibo Feng
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Wang Zhao
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Zhongyan Huang
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Zhao Yang
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| |
Collapse
|
11
|
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
|
12
|
High expression of GALNT7 promotes invasion and proliferation of glioma cells. Oncol Lett 2018; 16:6307-6314. [PMID: 30405766 PMCID: PMC6202485 DOI: 10.3892/ol.2018.9498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/20/2018] [Indexed: 01/06/2023] Open
Abstract
Polypeptide-N-acetyl-galactosaminlytransferase 7 (GALNT7), a member of the GalNAc-transferase family, has not been previously evaluated as a prognostic factor of glioblastoma (GBM) or low-grade glioma (LGG). Based on The Cancer Genome Atlas database and bioinformatics analyses, the expression of GALNT7 was demosntrated to be higher in GBM and LGG tissues than in normal brain tissue. The expression levels of GANLT7 were associated with age, tumor grade, survival rate, disease-free survival time and overall survival time. Gene correlation and gene-set enrichment analyses suggested that GALNT7 may affect the proliferative and invasive abilities of glioma cells through multiple signaling pathways, including regulation of the actin cytoskeleton, natural killer cell-mediated cytotoxicity, the janus kinase-signal transducer and activator of transcription (STAT) signaling pathway, cell adhesion molecules and extracellular matrix receptor interaction pathways. Furthermore, 5 target genes of GALNT7 involved in these signaling pathways were identified, including Crk, Rac family small GTPase 1, STAT3, poliovirus receptor and Tenascin C. In summary, high expression of GALNT7 was associated with poor prognosis of glioma, and may be used as an effective biomarker of glioma.
Collapse
|
13
|
Zakiryanova GK, Wheeler S, Shurin MR. Oncogenes in immune cells as potential therapeutic targets. Immunotargets Ther 2018; 7:21-28. [PMID: 29692982 PMCID: PMC5903485 DOI: 10.2147/itt.s150586] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The role of deregulated expression of oncogenes and tumor-suppressor genes in tumor development has been intensively investigated for decades. However, expression of oncogenes and their potential role in immune cell defects during carcinogenesis and tumor progression have not been thoroughly assessed. The defects in proto-oncogenes have been well documented and evaluated mostly in tumor cells, despite the fact that proto-oncogenes are expressed in all cells, including cells of the immune system. In this review, key studies from immune-mediated diseases that may be associated with oncogene signaling pathways are refocused to provide groundwork for beginning to understand the effects of oncogenes in and on the cancer-related immune system dysfunction.
Collapse
Affiliation(s)
- Gulnur K Zakiryanova
- Department Biophysics and Biomedicine, Faculty Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Sarah Wheeler
- Division of Clinical Immunopathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Michael R Shurin
- Division of Clinical Immunopathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Tajul Arifin K, Sulaiman S, Md Saad S, Ahmad Damanhuri H, Wan Ngah WZ, Mohd Yusof YA. Elevation of tumour markers TGF-β, M 2-PK, OV-6 and AFP in hepatocellular carcinoma (HCC)-induced rats and their suppression by microalgae Chlorella vulgaris. BMC Cancer 2017; 17:879. [PMID: 29268718 PMCID: PMC5740965 DOI: 10.1186/s12885-017-3883-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 12/07/2017] [Indexed: 12/24/2022] Open
Abstract
Background Chlorella vulgaris (ChV), a unicellular green algae has been reported to have anticancer and antioxidant effects. The aim of this study was to determine the chemopreventive effect of ChV on liver cancer induced rats by determining the level and expression of several liver tumour markers. Methods Male Wistar rats (200–250 g) were divided into 4 groups according to the diet given: control group (normal diet), ChV group with three different doses (50, 150 and 300 mg/kg body weight), liver cancer- induced group (choline deficient diet + 0.1% ethionine in drinking water or CDE group), and the treatment group (CDE group treated with three different doses of ChV). Rats were killed at 0, 4, 8 and 12 weeks of experiment and blood and tissue samples were taken from all groups for the determination of tumour markers expression alpha-fetoprotein (AFP), transforming growth factor-β (TGF-β), M2-pyruvate kinase (M2-PK) and specific antigen for oval cells (OV-6). Results Serum level of TGF-β increased significantly (p < 0.05) in CDE rats. However, ChV at all doses managed to decrease (p < 0.05) its levels to control values. Expressions of liver tumour markers AFP, TGF-β, M2-PK and OV-6 were significantly higher (p < 0.05) in tissues of CDE rats when compared to control showing an increased number of cancer cells during hepatocarcinogenesis. ChV at all doses reduced their expressions significantly (p < 0.05). Conclusions Chlorella vulgaris has chemopreventive effect by downregulating the expression of tumour markers M2-PK, OV-6, AFP and TGF-β, in HCC-induced rats.
Collapse
Affiliation(s)
- Khaizurin Tajul Arifin
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Suhaniza Sulaiman
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Suhana Md Saad
- Department of Diagnostic & Allied Health Sciences, Faculty of Health & Life Sciences, Management & Science University (MSU), University Drive, Seksyen 13, 40100, Shah Alam, Selangor, Malaysia
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Wan Zurinah Wan Ngah
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Yasmin Anum Mohd Yusof
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, Malaysia.
| |
Collapse
|
15
|
Immunological Evasion in Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7487313. [PMID: 27294132 PMCID: PMC4884578 DOI: 10.1155/2016/7487313] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 04/19/2016] [Indexed: 12/25/2022]
Abstract
Glioblastoma is the most aggressive tumor in Central Nervous System in adults. Among its features, modulation of immune system stands out. Although immune system is capable of detecting and eliminating tumor cells mainly by cytotoxic T and NK cells, tumor microenvironment suppresses an effective response through recruitment of modulator cells such as regulatory T cells, monocyte-derived suppressor cells, M2 macrophages, and microglia as well as secretion of immunomodulators including IL-6, IL-10, CSF-1, TGF-β, and CCL2. Other mechanisms that induce immunosuppression include enzymes as indolamine 2,3-dioxygenase. For this reason it is important to develop new therapies that avoid this immune evasion to promote an effective response against glioblastoma.
Collapse
|
16
|
Salazar-Ramiro A, Ramírez-Ortega D, Pérez de la Cruz V, Hérnandez-Pedro NY, González-Esquivel DF, Sotelo J, Pineda B. Role of Redox Status in Development of Glioblastoma. Front Immunol 2016; 7:156. [PMID: 27199982 PMCID: PMC4844613 DOI: 10.3389/fimmu.2016.00156] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/11/2016] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive neoplasia, prognosis remains dismal, and current therapy is mostly palliative. There are no known risk factors associated with gliomagenesis; however, it is well established that chronic inflammation in brain tissue induces oxidative stress in astrocytes and microglia. High quantities of reactive species of oxygen into the cells can react with several macromolecules, including chromosomal and mitochondrial DNA, leading to damage and malfunction of DNA repair enzymes. These changes bring genetic instability and abnormal metabolic processes, favoring oxidative environment and increase rate of cell proliferation. In GBM, a high metabolic rate and increased basal levels of reactive oxygen species play an important role as chemical mediators in the regulation of signal transduction, protecting malignant cells from apoptosis, thus creating an immunosuppressive environment. New redox therapeutics could reduce oxidative stress preventing cellular damage and high mutation rate accompanied by chromosomal instability, reducing the immunosuppressive environment. In addition, therapies directed to modulate redox rate reduce resistance and moderate the high rate of cell proliferation, favoring apoptosis of tumoral cells. This review describes the redox status in GBM, and how this imbalance could promote gliomagenesis through genomic and mitochondrial DNA damage, inducing the pro-oxidant and proinflammatory environment involved in tumor cell proliferation, resistance, and immune escape. In addition, some therapeutic agents that modulate redox status and might be advantageous in therapy against GBM are described.
Collapse
Affiliation(s)
- Aleli Salazar-Ramiro
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | - Daniela Ramírez-Ortega
- Neurochemistry Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | | | | | | | - Julio Sotelo
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | - Benjamín Pineda
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| |
Collapse
|