1
|
Wang L, He Z, Fan S, Mo L, Li Y, Yuan X, Xu B, Mou Y, Yin Y. Quantitative analysis of immune cells within the tumor microenvironment of glioblastoma and their relevance for prognosis. Int Immunopharmacol 2024; 142:113109. [PMID: 39255678 DOI: 10.1016/j.intimp.2024.113109] [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: 06/20/2024] [Revised: 08/09/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
Glioblastoma (GBM) is a high malignant tumor with no effective treatment. To comprehensively characterize the landscape of immune cells in GBM and evaluate their correlation with prognosis, we developed a multispectral fluorescent imaging pipeline that included tumor-infiltrating lymphocytic markers (CD3, CD4, CD8, FOXP3, NKP46), immune checkpoint markers (PD-1, PD-L1), and markers to characterize myeloid cells (CD68, CD66b, CD163, HLA-DR), to spatially quantify 18 immune cell subsets in 21 GBM cases. We found that macrophages are the most abundant in GBM microenvironment, followed by T cells and neutrophils, while NK and NKT cells are the least. Previously unreported CD8+ Treg, PD-L1+ neutrophils, and high proportion of PD-1+ NK and PD-1+ T cells were also detected. Single high densities of PD-1+CD8+ T cells, neutrophils, and PD-L1-expressing CD68+ cells were associated with longer survival. Moreover, closer proximity of T cells to PD-L1+ macrophages or PD-L1+ neutrophils were associated with poor prognosis. Correlative analysis revealed circulating PMN-MDSC and e-MDSC were positively correlated with intratumoral M2 macrophages, while circulating NK cells were inversely associated with infiltrating CD4+ Treg cells in GBM patients. Our findings highlighted the potential roles of infiltrating immune cells in prognosis prediction and developing novel immunotherapeutic strategies for GBM patients.
Collapse
Affiliation(s)
- Lu Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Zhenqiang He
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuning Fan
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Li Mo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Yanhui Yin
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China.
| |
Collapse
|
2
|
Wu J, Wang N. Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review). Mol Med Rep 2024; 30:221. [PMID: 39364736 PMCID: PMC11462401 DOI: 10.3892/mmr.2024.13344] [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: 02/17/2023] [Accepted: 11/11/2023] [Indexed: 10/05/2024] Open
Abstract
Glioblastoma (GBM) is the most common central nervous system malignancy in adults. GBM may be classified as grade IV diffuse astrocytoma according to the 2021 World Health Organization revised classification of central nervous system tumors, which means it is the most aggressive, invasive, undifferentiated type of tumor. Immune checkpoint blockade (ICB), particularly anti‑programmed cell death protein‑1 (PD‑1)/PD‑1 ligand‑1 immunotherapy, has been confirmed to be successful across several tumor types. However, in GBM, this treatment is still uncommon and the efficacy is unpredictable, and <10% of patients show long‑term responses. Recently, numerous studies have been conducted to explore what factors may indicate or affect the ICB response rate in GBM, including molecular alterations, immune expression signatures and immune infiltration. The present review aimed to summarize the current progress to improve the understanding of immunotherapy for GBM.
Collapse
Affiliation(s)
- Jianheng Wu
- Department of Neurosurgery, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Nannan Wang
- Department of Gastroenterology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| |
Collapse
|
3
|
Cheng SL, Lee HM, Li CP, Lin MW, Chou MY, Yen YT, Wu TH, Lian YC, Shih YC, Chiang CS, Chen TW, Wan D, Chen Y. Robust and Sustained STING Pathway Activation via Hydrogel-Based In Situ Vaccination for Cancer Immunotherapy. ACS NANO 2024; 18:29439-29456. [PMID: 39405469 PMCID: PMC11526424 DOI: 10.1021/acsnano.3c12337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
The stimulator of interferon genes (STING) pathway is crucial for tumor immunity, leading to the exploration of STING agonists as potential immunotherapy adjuvants. However, their clinical application faces obstacles including poor pharmacokinetics, transient activation, and an immunosuppressive tumor microenvironment (TME). Addressing these limitations, our study aims to develop an injectable silk fibroin hydrogel-based in situ vaccine. It incorporates a nanoscale STING agonist, an immunogenic cell death (ICD) inducer, and an immunomodulator to ensure their controlled and sustained release. cGAMP nanoparticles (cGAMPnps) with a core-shell structure ensure optimal delivery of cGAMP to dendritic cells (DCs), thereby activating the STING pathway and fostering DC maturation. ICD-associated damage-associated molecular patterns amplify and prolong STING activation via enhanced type I IFN and other inflammatory pathways, along with delayed degradation of cGAMP and STING. Furthermore, the STING-driven vascular normalization by cGAMPnps and ICD, in conjunction with immunomodulators like antiprogrammed cell death protein 1 antibody (anti-PD-1 Ab) or OX40 ligand (OX40L), effectively remodels the immunosuppressive TME. This in situ gel vaccine, when used independently or with surgery as neoadjuvant/adjuvant immunotherapy, enhances DC and CD8+ T-cell activation, suppressing tumor progression and recurrence across various immunologically cold tumor models. It revolutionizes the application of STING agonists in cancer immunotherapy, offering substantial promise for improving outcomes across a broad spectrum of malignancies.
Collapse
Affiliation(s)
- Sheng-Liang Cheng
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
- International
Intercollegiate Ph.D. Program, National
Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hsin-Mei Lee
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
| | - Chung-Pin Li
- Division
of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Division
of Clinical Skills Training, Department of Medical Education, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Therapeutic
and Research Center of Pancreatic Cancer, Veterans General Hospital, Taipei 11217, Taiwan
- School
of
Medicine, College of Medicine, National
Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Mei-Wei Lin
- Biomedical
Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 310401, Taiwan
| | - Min-Yuan Chou
- Biomedical
Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 310401, Taiwan
| | - Yu-Ting Yen
- Institute
of Translational Medicine and New Drug Development, School of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Tun-Han Wu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
| | - Yun-Chen Lian
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
| | - Yu-Chuan Shih
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
| | - Chi-Shiun Chiang
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ting-Wen Chen
- Institute
of Bioinformatics and Systems Biology, National
Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent
Drug Systems and Smart Bio-devices, National
Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
| | - Dehui Wan
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
| | - Yunching Chen
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, National Tsing
Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
4
|
Zhang P, Li C, Wang Y, Chi X, Sun T, Zhang Q, Zhang Y, Ji N. Expression features of targets for anti-glioma CAR-T cell immunotherapy. J Neurooncol 2024:10.1007/s11060-024-04855-4. [PMID: 39467936 DOI: 10.1007/s11060-024-04855-4] [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: 07/31/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE To investigate the expression features of common anti-glioma CAR-T targets (B7H3, CSPG4, EGFRv III, HER2 and IL-13Ra2) in gliomas with different grades and molecular subtypes, and explore the association of target expression with glioma malignant or immune phenotypes including immune evasion, stemness, antigen presentation, and tumor angiogenesis. METHODS Opal™ Multiplex immunofluorescence staining was performed on glioma tissues to detect the expression of targets, and biomarkers related to the phenotypes. RESULTS High variety of CAR-T target expression among glioma subtypes was observed. GBMs exhibited the highest expression level of all the examined targets among glioma subtypes. In all glioma cases, CSPG4 was the most prevalent target covering over 84% glioma cases, followed by B7H3 at over 64%. B7H3 exhibited the highest coverage (94%) in GBMs while CSPG4 was the most popular target in both oligodendrogliomas and astrocytomas, covering 94% and 80% cases, respectively. Bi or tri-target combination strategies markedly expanded the tumor coverage across glioma cases while increased tumor-cell coverage within tumor. PD-L1 expression was significantly enriched in all the target-positive cells (except the EGFRvIII+ cells); CD133 expression was higher in the CSPG4+ or IL-13Ra2+ cells, and CD31 elevated in the B7H3+ cells, as compared with their negative cell populations. CONCLUSION Anti-glioma CAR-T targets have heterogenous expression and distinct tumor coverage among glioma subtypes, and closely correlate with glioma malignant or immune phenotypes.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Chunzhao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiaohan Chi
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tai Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Qianhe Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yang Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nan Si Huan Xi Lu 119, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China.
| |
Collapse
|
5
|
Ahmady F, Curpen P, Perriman L, Fonseca Teixeira A, Wu S, Zhu HJ, Poddar A, Jayachandran A, Kannourakis G, Luwor RB. Reduced T and NK Cell Activity in Glioblastoma Patients Correlates with TIM-3 and BAT3 Dysregulation. Cells 2024; 13:1777. [PMID: 39513882 PMCID: PMC11545661 DOI: 10.3390/cells13211777] [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: 08/22/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Inhibitory receptors are critical for regulating immune cell function. In cancer, these receptors are often over-expressed on the cell surface of T and NK cells, leading to reduced anti-tumor activity. Here, through the analysis of 11 commonly studied checkpoint and inhibitory receptors, we discern that only HAVCR2 (TIM3) and ENTPD1 (CD39) display significantly greater gene expression in glioblastoma compared to normal brain and lower grade glioma. Cell surface TIM-3, but not ENTPD1, was also elevated on activated CD4+ and CD8+ T cells, as well as on NK cells from glioblastoma patients compared to healthy donor T and NK cells. A subsequent analysis of molecules known to co-ordinate TIM-3 function and regulation was performed, which revealed that BAT3 expression was significantly reduced in CD4+ and CD8+ T cells, as well as NK cells from glioblastoma patients compared to counterparts from healthy donors. These pro-inhibitory changes are also correlated with reduced levels of the activation marker CD69 and the pro-inflammatory cytokine IFNγ in CD4+ and CD8+ T cells, as well as NK cells from glioblastoma patients. Collectively, these data reveal that glioblastoma-mediated CD4+ and CD8+ T cell and NK cell suppression is due, at least in part, to dysregulated TIM-3 and BAT3 expression and the associated downstream immunoregulatory and dysfunctional effects.
Collapse
Affiliation(s)
- Farah Ahmady
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Peter Curpen
- Townsville Hospital and Health Service, James Cook University, Townsville, QLD 4814, Australia;
| | - Louis Perriman
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Adilson Fonseca Teixeira
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| | - Siqi Wu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| | - Hong-Jian Zhu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| | - Arpita Poddar
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Rodney B. Luwor
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (L.P.); (A.P.); (A.J.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3050, Australia; (A.F.T.); (S.W.); (H.-J.Z.)
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| |
Collapse
|
6
|
Malacopol AT, Holst PJ. Cancer Vaccines: Recent Insights and Future Directions. Int J Mol Sci 2024; 25:11256. [PMID: 39457036 PMCID: PMC11508577 DOI: 10.3390/ijms252011256] [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: 09/11/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The field of cancer immunotherapy has seen incredible advancements in the past decades. mRNA-based cancer vaccines generating de novo T cell responses, particularly against tumor-specific antigens (TSAs), have demonstrated promising clinical outcomes and overcome diverse challenges. Despite the high potential of neoantigens to provide personalized immunotherapies through their tumor specificity and immunogenicity, challenges related to the scarcity of immunogenic neoepitopes have prompted continuous research towards finding new tumor-associated antigens (TAAs) and broader therapeutic frameworks, which may now learn from the genuine successes obtained with neoantigens. As an example, human endogenous retroviruses (HERVs) have emerged as potential alternatives to tumor neoantigens due to their high tumoral expression and ability to elicit both T cell reactivity and B cell responses associated with the efficacy of existing immunotherapies. This review aims to assess the status and limitations of TSA-directed mRNA cancer vaccines and the lessons that can be derived from these and checkpoint inhibitor studies to guide TAA vaccine development. We expect that shared B cell, CD4 and CD8 T cell antigen presentation will be key to stimulate continuous T cell expansion and efficacy for tumors that do not contain pre-existing tertiary lymphoid structures. When these structures are present in highly mutated tumors, the current checkpoint-based immunotherapies show efficacy even in immune privileged sites, and vaccines may hold the key to broaden efficacy to more tumor types and stages.
Collapse
Affiliation(s)
- Aretia-Teodora Malacopol
- HERVOLUTION Therapeutics, Copenhagen Bio Science (COBIS), 215 Nordre Fasanvej, DK2200 Copenhagen, Denmark;
| | - Peter Johannes Holst
- HERVOLUTION Therapeutics, Copenhagen Bio Science (COBIS), 215 Nordre Fasanvej, DK2200 Copenhagen, Denmark;
- Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, DK2200 Copenhagen, Denmark
| |
Collapse
|
7
|
Zheng Y, Chen X, Wang Y, Chen Z, Wu D. Phenolic-enabled nanotechnology: a new strategy for central nervous system disease therapy. J Zhejiang Univ Sci B 2024; 25:890-913. [PMID: 39420524 PMCID: PMC11494163 DOI: 10.1631/jzus.b2300839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 10/19/2024]
Abstract
Polyphenolic compounds have received tremendous attention in biomedicine because of their good biocompatibility and unique physicochemical properties. In recent years, phenolic-enabled nanotechnology (PEN) has become a hotspot of research in the medical field, and many promising studies have been reported, especially in the application of central nervous system (CNS) diseases. Polyphenolic compounds have superior anti-inflammatory and antioxidant properties, and can easily cross the blood‒brain barrier, as well as protect the nervous system from metabolic damage and promote learning and cognitive functions. However, although great advances have been made in this field, a comprehensive review regarding PEN-based nanomaterials for CNS therapy is lacking. A systematic summary of the basic mechanisms and synthetic strategies of PEN-based nanomaterials is beneficial for meeting the demand for the further development of novel treatments for CNS diseases. This review systematically introduces the fundamental physicochemical properties of PEN-based nanomaterials and their applications in the treatment of CNS diseases. We first describe the different ways in which polyphenols interact with other substances to form high-quality products with controlled sizes, shapes, compositions, and surface chemistry and functions. The application of PEN-based nanomaterials in the treatment of CNS diseases is then described, which provides a reference for subsequent research on the treatment of CNS diseases.
Collapse
Affiliation(s)
- Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. ,
- Zhejiang Rehabilitation Medical Center, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310009, China. ,
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. ,
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
8
|
Wan F, Li Y, Zhu J, Yu D, Liu H, Hu B. Exploring the prognostic value and potential therapeutic strategies of MS4A6A in glioblastoma: A comprehensive analysis of single-cell and multi-omics data. J Cell Mol Med 2024; 28:e70177. [PMID: 39470579 PMCID: PMC11520442 DOI: 10.1111/jcmm.70177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/30/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and treatment-resistant malignancy that poses a significant challenge in modern medicine. Despite advances in surgical resection, radiotherapy and chemotherapy, complete eradication of GBM remains elusive due to its diffuse invasion into the brain parenchyma and propensity for recurrence. The tumour microenvironment (TME), particularly macrophages, has emerged as a critical player in GBM progression, invasion and metastasis. In the immune microenvironment of glioma, MS4A6A exhibits unique expression characteristics in macrophages. This study aimed to investigate the potential role of MS4A6A, a gene associated with aging and neurodegenerative diseases, in GBM and its potential as a prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Fangchao Wan
- Department of Neurology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Yanling Li
- Department of Neurology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Jianming Zhu
- Department of Neurology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Dandan Yu
- Department of Electrocardiogram, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Hongjuan Liu
- Department of Neurology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| | - Bohong Hu
- Department of Neurology, Changde Hospital, Xiangya School of MedicineCentral South UniversityChangdeHunanChina
| |
Collapse
|
9
|
Tapescu I, Madsen PJ, Lowenstein PR, Castro MG, Bagley SJ, Fan Y, Brem S. The transformative potential of mRNA vaccines for glioblastoma and human cancer: technological advances and translation to clinical trials. Front Oncol 2024; 14:1454370. [PMID: 39399167 PMCID: PMC11466887 DOI: 10.3389/fonc.2024.1454370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Originally devised for cancer control, mRNA vaccines have risen to the forefront of medicine as effective instruments for control of infectious disease, notably their pivotal role in combating the COVID-19 pandemic. This review focuses on fundamental aspects of the development of mRNA vaccines, e.g., tumor antigens, vector design, and precise delivery methodologies, - highlighting key technological advances. The recent, promising success of personalized mRNA vaccines against pancreatic cancer and melanoma illustrates the potential value for other intractable, immunologically resistant, solid tumors, such as glioblastoma, as well as the potential for synergies with a combinatorial, immunotherapeutic approach. The impact and progress in human cancer, including pancreatic cancer, head and neck cancer, bladder cancer are reviewed, as are lessons learned from first-in-human CAR-T cell, DNA and dendritic cell vaccines targeting glioblastoma. Going forward, a roadmap is provided for the transformative potential of mRNA vaccines to advance cancer immunotherapy, with a particular focus on the opportunities and challenges of glioblastoma. The current landscape of glioblastoma immunotherapy and gene therapy is reviewed with an eye to combinatorial approaches harnessing RNA science. Preliminary preclinical and clinical data supports the concept that mRNA vaccines could be a viable, novel approach to prolong survival in patients with glioblastoma.
Collapse
Affiliation(s)
- Iulia Tapescu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peter J. Madsen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Pedro R. Lowenstein
- Department of Neurosurgery, The University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, The University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
| | - Stephen J. Bagley
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Yi Fan
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Steven Brem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
10
|
Dadario NB, Boyett DM, Teasley DE, Chabot PJ, Winans NJ, Argenziano MG, Sperring CP, Canoll P, Bruce JN. Unveiling the Inflammatory Landscape of Recurrent Glioblastoma through Histological-Based Assessments. Cancers (Basel) 2024; 16:3283. [PMID: 39409905 PMCID: PMC11476027 DOI: 10.3390/cancers16193283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
The glioblastoma (GBM) tumor microenvironment consists of a heterogeneous mixture of neoplastic and non-neoplastic cells, including immune cells. Tumor recurrence following standard-of-care therapy results in a rich landscape of inflammatory cells throughout the glioma-infiltrated cortex. Immune cells consisting of glioma-associated macrophages and microglia (GAMMs) overwhelmingly constitute the bulk of the recurrent glioblastoma (rGBM) microenvironment, in comparison to the highly cellular and proliferative tumor microenvironment characteristic of primary GBM. These immune cells dynamically interact within the tumor microenvironment and can contribute to disease progression and therapy resistance while also providing novel targets for emerging immunotherapies. Within these varying contexts, histological-based assessments of immune cells in rGBM, including immunohistochemistry (IHC) and immunofluorescence (IF), offer a critical way to visualize and examine the inflammatory landscape. Here, we exhaustively review the available body of literature on the inflammatory landscape in rGBM as identified through histological-based assessments. We highlight the heterogeneity of immune cells throughout the glioma-infiltrated cortex with a focus on microglia and macrophages, drawing insights from canonical and novel immune-cell histological markers to estimate cell phenotypes and function. Lastly, we discuss opportunities for immunomodulatory treatments aiming to harness the inflammatory landscape in rGBM.
Collapse
Affiliation(s)
- Nicholas B. Dadario
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Deborah M. Boyett
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Damian E. Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Peter J. Chabot
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Nathan J. Winans
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Michael G. Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Colin P. Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| |
Collapse
|
11
|
Keane L, Cryan JF, Gleeson JP. Exploiting the gut microbiome for brain tumour treatment. Trends Mol Med 2024:S1471-4914(24)00222-3. [PMID: 39256110 DOI: 10.1016/j.molmed.2024.08.008] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Increasing evidence suggests that the gut microbiome plays a key role in a host of pathological conditions, including cancer. Indeed, the bidirectional communication that occurs between the gut and the brain, known as the 'gut-brain axis,' has recently been implicated in brain tumour pathology. Here, we focus on current research that supports a gut microbiome-brain tumour link with emphasis on high-grade gliomas, the most aggressive of all brain tumours, and the impact on the glioma tumour microenvironment. We discuss the potential use of gut-brain axis signals to improve responses to current and future therapeutic approaches. We highlight that the success of novel treatment strategies may rely on patient-specific microbiome profiles, and these should be considered for personalised treatment approaches.
Collapse
Affiliation(s)
- Lily Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Jack P Gleeson
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland; CUH/UCC Cancer Centre, Cork University Hospital, Cork, Ireland.
| |
Collapse
|
12
|
Lastra Romero A, Seitz T, Zisiadis GA, Jeffery H, Osman AM. EDA2R reflects the acute brain response to cranial irradiation in liquid biopsies. Neuro Oncol 2024; 26:1617-1627. [PMID: 38683135 PMCID: PMC11376461 DOI: 10.1093/neuonc/noae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Cranial radiotherapy is standard of care for high-grade brain tumors and metastases; however, it induces debilitating neurocognitive impairments in cancer survivors, especially children. As the numbers of pediatric brain cancer survivors continue improving, the numbers of individuals developing life-long neurocognitive sequalae are consequently expected to rise. Yet, there are no established biomarkers estimating the degree of the irradiation-induced brain injury at completion of radiotherapy to predict the severity of the expected neurocognitive complications. We aimed to identify sensitive biomarkers associated with brain response to irradiation that can be measured in easily accessible clinical materials, such as liquid biopsies. METHODS Juvenile mice were subjected to cranial irradiation with 0.5, 1, 2, 4, and 8 Gy. Cerebrospinal fluid (CSF), plasma, and brains were collected at acute, subacute, and subchronic phases after irradiation, and processed for proteomic screens, and molecular and histological analyses. RESULTS We found that the levels of ectodysplasin A2 receptor (EDA2R), member of tumor necrosis factor receptor superfamily, increased significantly in the CSF after cranial irradiation, even at lower irradiation doses. The levels of EDA2R were increased globally in the brain acutely after irradiation and decreased over time. EDA2R was predominantly expressed by neurons, and the temporal dynamics of EDA2R in the brain was reflected in the plasma samples. CONCLUSIONS We propose EDA2R as a promising potential biomarker reflecting irradiation-induced brain injury in liquid biopsies. The levels of EDA2R upon completion of radiotherapy may aid in predicting the severity of IR-induced neurocognitive sequalae at a very early stage after treatment.
Collapse
Affiliation(s)
| | - Thea Seitz
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | | | - Holli Jeffery
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ahmed M Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Frąk M, Grenda A, Krawczyk P, Kuźnar-Kamińska B, Pazdrowski P, Kędra K, Chmielewska I, Milanowski J. The influence of nutritional status, lipid profile, leptin concentration and polymorphism of genes encoding leptin and neuropeptide Y on the effectiveness of immunotherapy in advanced NSCLC patients. BMC Cancer 2024; 24:937. [PMID: 39090596 PMCID: PMC11295594 DOI: 10.1186/s12885-024-12716-6] [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: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION Neuropeptide Y is a neurotransmitter in the nervous system and belongs to the orexigenic system that increases appetite. Its excessive secretion leads to obesity. Leptin is a pro-inflammatory adipokine (produced in adipose tissue) induced in obesity and may mediate increased antitumor immunity in obesity (including the promotion of M1 macrophages). Leptin and neuropeptide Y gene polymorphisms, causing increased leptin levels and the occurrence of obesity, and lipid profile disorders, may increase the effectiveness of immunotherapy. MATERIALS AND METHODS In 121 patients with advanced NSCLC without mutations in the EGFR gene and rearrangements of the ALK and ROS1 genes, undergoing immunotherapy (1st and 2nd line of treatment) or chemoimmunotherapy (1st line of treatment), we assessed BMI, lipid profile, PD-L1 expression on cancer cells using the immunohistochemical method (clone SP263 antibody), leptin concentration in blood serum by ELISA, polymorphisms in the promoter region of the genes for leptin (LEP) and neuropeptide Y (NPY) by real-time PCR. RESULTS Leptin concentration was significantly higher in obese patients than in patients with normal or low weight (p = 0.00003) and in patients with disease stabilization compared to patients with progression observed during immunotherapy (p = 0.012). Disease control occurred significantly more often in patients with the GA or AA genotype than patients with the GG genotype in the rs779039 polymorphism of the LEP gene. The median PFS in the entire study group was five months (95% CI: 3-5.5), and the median OS was 12 months (95% CI: 8-16). Median PFS was highest in patients with TPS ≥ 50% (6.5 months) and in obese patients (6.6 months). Obese patients also had a slightly longer median OS compared to other patients (23.8 vs. 13 months). The multivariate Cox logistic regression test showed that the only factor reducing the risk of progression was TPS ≥ 50% (HR = 0.6068, 95% CI: 0.4001-0.9204, p = 0, 0187), and the only factor reducing the risk of death was high leptin concentration (HR = 0.6743, 95% CI: 0.4243-1.0715, p = 0.0953). CONCLUSION Assessment of nutritional status, serum leptin concentration and polymorphisms in the LEP gene may be of additional importance in predicting the effectiveness of immunotherapy and chemoimmunotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Małgorzata Frąk
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Barbara Kuźnar-Kamińska
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Paweł Pazdrowski
- Department of Head, Neck Surgery and Laryngological Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Karolina Kędra
- Institute of Physical Chemistry, Polish Academy of Sciences in Warsaw, Warsaw, Poland
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| |
Collapse
|
14
|
Yang X, Niu W, Wu K, Li X, Hou H, Tan Y, Wang X, Yang G, Wang L, Zhang H. Diffusion kurtosis imaging-based habitat analysis identifies high-risk molecular subtypes and heterogeneity matching in diffuse gliomas. Ann Clin Transl Neurol 2024; 11:2073-2087. [PMID: 38887966 PMCID: PMC11330218 DOI: 10.1002/acn3.52128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/14/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE High-risk types of diffuse gliomas in adults include isocitrate dehydrogenase (IDH) wild-type glioblastomas and grade 4 astrocytomas. Achieving noninvasive prediction of high-risk molecular subtypes of gliomas is important for personalized and precise diagnosis and treatment. METHODS We retrospectively collected data from 116 patients diagnosed with adult diffuse gliomas. Multiple high-risk molecular markers were tested, and various habitat models and whole-tumor models were constructed based on preoperative routine and diffusion kurtosis imaging (DKI) sequences to predict high-risk molecular subtypes of gliomas. Feature selection and model construction utilized Least absolute shrinkage and selection operator (LASSO) and support vector machine (SVM). Finally, the Wilcoxon rank-sum test was employed to explore the correlation between habitat quantitative features (intra-tumor heterogeneity score,ITH score) and heterogeneity, as well as high-risk molecular subtypes. RESULTS The results showed that the habitat analysis model based on DKI performed remarkably well (with AUC values reaching 0.977 and 0.902 in the training and test sets, respectively). The model's performance was further enhanced when combined with clinical variables. (The AUC values were 0.994 and 0.920, respectively.) Additionally, we found a close correlation between ITH score and heterogeneity, with statistically significant differences observed between high-risk and non-high-risk molecular subtypes. INTERPRETATION The habitat model based on DKI is an ideal means for preoperatively predicting high-risk molecular subtypes of gliomas, holding significant value for noninvasively alerting malignant gliomas and those with malignant transformation potential.
Collapse
Affiliation(s)
- Xiangli Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalTaiyuan030032China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Wenju Niu
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Kai Wu
- Department of Information ManagementFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiang Li
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Heng Hou
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Yan Tan
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiaochun Wang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Guoqiang Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Lei Wang
- Beijing Tiantan HospitalCapital Medical UniversityBeijing100050China
| | - Hui Zhang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Intelligent Imaging Big Data and Functional Nano‐imaging Engineering Research Center of Shanxi ProvinceFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
15
|
Seyhan AA. Circulating Liquid Biopsy Biomarkers in Glioblastoma: Advances and Challenges. Int J Mol Sci 2024; 25:7974. [PMID: 39063215 PMCID: PMC11277426 DOI: 10.3390/ijms25147974] [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: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Gliomas, particularly glioblastoma (GBM), represent the most prevalent and aggressive tumors of the central nervous system (CNS). Despite recent treatment advancements, patient survival rates remain low. The diagnosis of GBM traditionally relies on neuroimaging methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans and postoperative confirmation via histopathological and molecular analysis. Imaging techniques struggle to differentiate between tumor progression and treatment-related changes, leading to potential misinterpretation and treatment delays. Similarly, tissue biopsies, while informative, are invasive and not suitable for monitoring ongoing treatments. These challenges have led to the emergence of liquid biopsy, particularly through blood samples, as a promising alternative for GBM diagnosis and monitoring. Presently, blood and cerebrospinal fluid (CSF) sampling offers a minimally invasive means of obtaining tumor-related information to guide therapy. The idea that blood or any biofluid tests can be used to screen many cancer types has huge potential. Tumors release various components into the bloodstream or other biofluids, including cell-free nucleic acids such as microRNAs (miRNAs), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), proteins, extracellular vesicles (EVs) or exosomes, metabolites, and other factors. These factors have been shown to cross the blood-brain barrier (BBB), presenting an opportunity for the minimally invasive monitoring of GBM as well as for the real-time assessment of distinct genetic, epigenetic, transcriptomic, proteomic, and metabolomic changes associated with brain tumors. Despite their potential, the clinical utility of liquid biopsy-based circulating biomarkers is somewhat constrained by limitations such as the absence of standardized methodologies for blood or CSF collection, analyte extraction, analysis methods, and small cohort sizes. Additionally, tissue biopsies offer more precise insights into tumor morphology and the microenvironment. Therefore, the objective of a liquid biopsy should be to complement and enhance the diagnostic accuracy and monitoring of GBM patients by providing additional information alongside traditional tissue biopsies. Moreover, utilizing a combination of diverse biomarker types may enhance clinical effectiveness compared to solely relying on one biomarker category, potentially improving diagnostic sensitivity and specificity and addressing some of the existing limitations associated with liquid biomarkers for GBM. This review presents an overview of the latest research on circulating biomarkers found in GBM blood or CSF samples, discusses their potential as diagnostic, predictive, and prognostic indicators, and discusses associated challenges and future perspectives.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
16
|
Coupey J, Leblond MM, Hue ES, Valable S. Flow cytometry detection and quantification of circulating leukocyte subpopulations in mice after brain irradiation. Methods Cell Biol 2024; 189:135-152. [PMID: 39393880 DOI: 10.1016/bs.mcb.2024.06.004] [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: 10/13/2024]
Abstract
In the context of high-grade gliomas such as glioblastoma (GBM), the immune part of the tumor microenvironment (TME) is involved in tumor growth and tumor recurrence. It is mostly represented by high amount of macrophages and low amount of lymphocytes. GBM in itself as well as x-ray-based radiotherapy, a standard treatment for brain tumors, are also associated with systemic effects like lymphopenia that correlates with a poor prognosis. This contributes to the immune-suppressive nature of the TME and may explain the lack of the anti-tumor immune response. Radiation-induced lymphopenia (RIL) is generally evaluated on CD4+ and CD8+ count or on a CBC (complete blood count), but the heterogeneity of the subtypes prompts us to explore them in detail to better understand the cellular response to brain irradiation. To facilitate and develop the evaluation of x-ray brain exposure on circulating immune cells, we developed a reproducible and reliable method to quantify the variation of lymphoid and myeloid subtypes using flow cytometry after brain irradiation in the rodent.
Collapse
Affiliation(s)
- Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France
| | - Marine M Leblond
- Department of Oncology UNIL CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Erika S Hue
- LABÉO, Saint-Contest, France; Normandie Université, UNICAEN BIOTARGEN, Saint-Contest, France
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP Cyceron, Caen, France.
| |
Collapse
|
17
|
Yu K, Tian Q, Feng S, Zhang Y, Cheng Z, Li M, Zhu H, He J, Li M, Xiong X. Integration analysis of cell division cycle-associated family genes revealed potential mechanisms of gliomagenesis and constructed an artificial intelligence-driven prognostic signature. Cell Signal 2024; 119:111168. [PMID: 38599441 DOI: 10.1016/j.cellsig.2024.111168] [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: 02/26/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Cell division cycle-associated (CDCA) gene family members are essential cell proliferation regulators and play critical roles in various cancers. However, the function of the CDCA family genes in gliomas remains unclear. This study aims to elucidate the role of CDCA family members in gliomas using in vitro and in vivo experiments and bioinformatic analyses. We included eight glioma cohorts in this study. An unsupervised clustering algorithm was used to identify novel CDCA gene family clusters. Then, we utilized multi-omics data to elucidate the prognostic disparities, biological functionalities, genomic alterations, and immune microenvironment among glioma patients. Subsequently, the scRNA-seq analysis and spatial transcriptomic sequencing analysis were carried out to explore the expression distribution of CDCA2 in glioma samples. In vivo and in vitro experiments were used to investigate the effects of CDCA2 on the viability, migration, and invasion of glioma cells. Finally, based on ten machine-learning algorithms, we constructed an artificial intelligence-driven CDCA gene family signature called the machine learning-based CDCA gene family score (MLCS). Our results suggested that patients with the higher expression levels of CDCA family genes had a worse prognosis, more activated RAS signaling pathways, and more activated immunosuppressive microenvironments. CDCA2 knockdown inhibited the proliferation, migration, and invasion of glioma cells. In addition, the MLCS had robust and favorable prognostic predictive ability and could predict the response to immunotherapy and chemotherapy drug sensitivity.
Collapse
Affiliation(s)
- Kai Yu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Shi Feng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Ziqi Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Mingyang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Jianying He
- Department of Orthopedics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi Province, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
18
|
Stergiopoulos GM, Concilio SC, Galanis E. An Update on the Clinical Status, Challenges, and Future Directions of Oncolytic Virotherapy for Malignant Gliomas. Curr Treat Options Oncol 2024; 25:952-991. [PMID: 38896326 DOI: 10.1007/s11864-024-01211-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 06/21/2024]
Abstract
OPINION STATEMENT Malignant gliomas are common central nervous system tumors that pose a significant clinical challenge due to the lack of effective treatments. Glioblastoma (GBM), a grade 4 malignant glioma, is the most prevalent primary malignant brain tumor and is associated with poor prognosis. Current clinical trials are exploring various strategies to combat GBM, with oncolytic viruses (OVs) appearing particularly promising. In addition to ongoing and recently completed clinical trials, one OV (Teserpaturev, Delytact®) received provisional approval for GBM treatment in Japan. OVs are designed to selectively target and eliminate cancer cells while promoting changes in the tumor microenvironment that can trigger and support long-lasting anti-tumor immunity. OVs offer the potential to remodel the tumor microenvironment and reverse systemic immune exhaustion. Additionally, an increasing number of OVs are armed with immunomodulatory payloads or combined with immunotherapy approaches in an effort to promote anti-tumor responses in a tumor-targeted manner. Recently completed oncolytic virotherapy trials can guide the way for future treatment individualization through patient preselection, enhancing the likelihood of achieving the highest possible clinical success. These trials also offer valuable insight into the numerous challenges inherent in malignant glioma treatment, some of which OVs can help overcome.
Collapse
Affiliation(s)
| | | | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
19
|
Ahmad I, Altameemi KKA, Hani MM, Ali AM, Shareef HK, Hassan ZF, Alubiady MHS, Al-Abdeen SHZ, Shakier HG, Redhee AH. Shifting cold to hot tumors by nanoparticle-loaded drugs and products. Clin Transl Oncol 2024:10.1007/s12094-024-03577-3. [PMID: 38922537 DOI: 10.1007/s12094-024-03577-3] [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: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Cold tumors lack antitumor immunity and are resistant to therapy, representing a major challenge in cancer medicine. Because of the immunosuppressive spirit of the tumor microenvironment (TME), this form of tumor has a low response to immunotherapy, radiotherapy, and also chemotherapy. Cold tumors have low infiltration of immune cells and a high expression of co-inhibitory molecules, such as immune checkpoints and immunosuppressive molecules. Therefore, targeting TME and remodeling immunity in cold tumors can improve the chance of tumor repression after therapy. However, tumor stroma prevents the infiltration of inflammatory cells and hinders the penetration of diverse molecules and drugs. Nanoparticles are an intriguing tool for the delivery of immune modulatory agents and shifting cold to hot tumors. In this review article, we discuss the mechanisms underlying the ability of nanoparticles loaded with different drugs and products to modulate TME and enhance immune cell infiltration. We also focus on newest progresses in the design and development of nanoparticle-based strategies for changing cold to hot tumors. These include the use of nanoparticles for targeted delivery of immunomodulatory agents, such as cytokines, small molecules, and checkpoint inhibitors, and for co-delivery of chemotherapy drugs and immunomodulatory agents. Furthermore, we discuss the potential of nanoparticles for enhancing the efficacy of cancer vaccines and cell therapy for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | - Mohaned Mohammed Hani
- Department of Medical Instrumentation Engineering Techniques, Imam Ja'afar Al-Sadiq University, Al Muthanna, Iraq
| | - Afaq Mahdi Ali
- Department of Pharmaceutics, Al-Turath University College, Baghdad, Iraq
| | - Hasanain Khaleel Shareef
- Department of Medical Biotechnology, College of Science, Al-Mustaqbal University, Hilla, Iraq
- Biology Department, College of Science for Women, University of Babylon, Hilla, Iraq
| | | | | | | | | | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
20
|
ZHU ZHAOYING, HU YANJIA, YE FENG, TENG HAIBO, YOU GUOLIANG, ZENG YUNHUI, TIAN MENG, XU JIANGUO, LI JIN, LIU ZHIYONG, LIU HAO, ZHENG NIANDONG. IKIP downregulates THBS1/FAK signaling to suppress migration and invasion by glioblastoma cells. Oncol Res 2024; 32:1173-1184. [PMID: 38948026 PMCID: PMC11211642 DOI: 10.32604/or.2024.042456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 02/04/2024] [Indexed: 07/02/2024] Open
Abstract
Background Inhibitor of NF-κB kinase-interacting protein (IKIP) is known to promote proliferation of glioblastoma (GBM) cells, but how it affects migration and invasion by those cells is unclear. Methods We compared levels of IKIP between glioma tissues and normal brain tissue in clinical samples and public databases. We examined the effects of IKIP overexpression and knockdown on the migration and invasion of GBM using transwell and wound healing assays, and we compared the transcriptomes under these different conditions to identify the molecular mechanisms involved. Results Based on data from our clinical samples and from public databases, IKIP was overexpressed in GBM tumors, and its expression level correlated inversely with survival. IKIP overexpression in GBM cells inhibited migration and invasion in transwell and wound healing assays, whereas IKIP knockdown exerted the opposite effects. IKIP overexpression in GBM cells that were injected into mouse brain promoted tumor growth but inhibited tumor invasion of surrounding tissue. The effects of IKIP were associated with downregulation of THBS1 mRNA and concomitant inhibition of THBS1/FAK signaling. Conclusions IKIP inhibits THBS1/FAK signaling to suppress migration and invasion of GBM cells.
Collapse
Affiliation(s)
- ZHAOYING ZHU
- Department of Neurosurgery, The Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - YANJIA HU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - FENG YE
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - HAIBO TENG
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - GUOLIANG YOU
- Department of Neurosurgery, The Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - YUNHUI ZENG
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - MENG TIAN
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - JIANGUO XU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - JIN LI
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - ZHIYONG LIU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - HAO LIU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - NIANDONG ZHENG
- Department of Neurosurgery, The Affiliated Hospital of Southwestern Medical University, Luzhou, China
| |
Collapse
|
21
|
Liu CC, Yang WB, Chien CH, Wu CL, Chuang JY, Chen PY, Chu JM, Cheng SM, Qiu LY, Chang YC, Hwang DY, Huang CY, Lee JS, Chang KY. CXCR7 activation evokes the anti-PD-L1 antibody against glioblastoma by remodeling CXCL12-mediated immunity. Cell Death Dis 2024; 15:434. [PMID: 38898023 PMCID: PMC11187218 DOI: 10.1038/s41419-024-06784-6] [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: 01/24/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
The interaction between glioblastoma cells and glioblastoma-associated macrophages (GAMs) influences the immunosuppressive tumor microenvironment, leading to ineffective immunotherapies. We hypothesized that disrupting the communication between tumors and macrophages would enhance the efficacy of immunotherapies. Transcriptomic analysis of recurrent glioblastoma specimens indicated an enhanced neuroinflammatory pathway, with CXCL12 emerging as the top-ranked gene in secretory molecules. Single-cell transcriptome profiling of naïve glioblastoma specimens revealed CXCL12 expression in tumor and myeloid clusters. An analysis of public glioblastoma datasets has confirmed the association of CXCL12 with disease and PD-L1 expression. In vitro studies have demonstrated that exogenous CXCL12 induces pro-tumorigenic characteristics in macrophage-like cells and upregulated PD-L1 expression through NF-κB signaling. We identified CXCR7, an atypical receptor for CXCL12 predominantly present in tumor cells, as a negative regulator of CXCL12 expression by interfering with extracellular signal-regulated kinase activation. CXCR7 knockdown in a glioblastoma mouse model resulted in worse survival outcomes, increased PD-L1 expression in GAMs, and reduced CD8+ T-cell infiltration compared with the control group. Ex vivo T-cell experiments demonstrated enhanced cytotoxicity against tumor cells with a selective CXCR7 agonist, VUF11207, reversing GAM-induced immunosuppression in a glioblastoma cell-macrophage-T-cell co-culture system. Notably, VUF11207 prolonged survival and potentiated the anti-tumor effect of the anti-PD-L1 antibody in glioblastoma-bearing mice. This effect was mitigated by an anti-CD8β antibody, indicating the synergistic effect of VUF11207. In conclusion, CXCL12 conferred immunosuppression mediated by pro-tumorigenic and PD-L1-expressing GAMs in glioblastoma. Targeted activation of glioblastoma-derived CXCR7 inhibits CXCL12, thereby eliciting anti-tumor immunity and enhancing the efficacy of anti-PD-L1 antibodies.
Collapse
Affiliation(s)
- Chan-Chuan Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Wen-Bin Yang
- Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Hung Chien
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Cheng-Lin Wu
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Jian-Ying Chuang
- Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan
- International Master Program in Medical Neuroscience, Taipei Medical University, Taipei, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pin-Yuan Chen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Jui-Mei Chu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Siao Muk Cheng
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Li-Ying Qiu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yung-Chieh Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- TMU Research Center of Cancer Translational Medicine; Taipei Cancer Center; Taipei Medical University Hospital, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Daw-Yang Hwang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chih-Yuan Huang
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jung-Shun Lee
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
22
|
Duan J, Chen J, Lin Y, Lin SL, Wu J. Endocannabinoid Receptor 2 Function is Associated with Tumor-Associated Macrophage Accumulation and Increases in T Cell Number to Initiate a Potent Antitumor Response in a Syngeneic Murine Model of Glioblastoma. Cannabis Cannabinoid Res 2024. [PMID: 38888628 DOI: 10.1089/can.2024.0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024] Open
Abstract
Introduction: Glioblastoma patients have a highly immunosuppressive tumor microenvironment and systemic immunosuppression that comprise a major barrier to immune checkpoint therapy. Based on the production of endocannabinoids by glioblastomas, we explored involvement of endocannabinoid receptor 2 (CB2R), encoded by the CNR2 gene, which is predominantly expressed by immune cells, in glioblastoma-related immunosuppression. Materials & Methods: Bioinformatics of human glioblastoma databases was used to correlate enzymes involved in the synthesis and degradation of endocannabinoids, as well as CB2Rs, with patient overall survival. Intrastriatal administration of luciferase-expressing, murine GL261 glioblastoma cells was used to establish in in vivo glioblastoma model for characterization of tumor growth and intratumoral immune cell infiltration, as well as provide immune cells for in vitro co-culture experiments. Involvement of CB2Rs was determined by treatment with CB2R agonist (GW405833) or CB2R antagonist (AM630). ELISA, FACS, and immunocytochemistry were used to determine perforin, granzyme B, and surface marker levels. Results: Bioinformatics of human glioblastoma databases showed high expression of CB2R and elevated endocannabinoid production correlated with poorer prognosis, and involved immune-associated pathways. AM630treatment of GL261 glioblastoma-bearing mice induced a potent antitumor response, with survival plateauing at 50% on Day 40, when all control mice (median survival 28 days) and mice treated with GW405833 (median survival 21 days) had died. Luciferase tumor imaging revealed accelerated tumor growth by GW405833 treatment, but stable or regressing tumors in AM630-treated mice. Notably, in spleens, AM630 treatment caused an 83% decrease in monocytes/macrophages, and 1.8- and 1.6-fold increases in CD8+ and CD4+ cells, respectively. Within tumors, there was a corresponding decrease in tumor-associated macrophages (TAMs) and increase in CD8+ T cells. In vitro, lymphocytes from AM630-treated mice showed greater cytotoxic function (increased percentage of perforin- and granzyme B-positive CD8+ T cells). Discussion: These results suggest that inhibition of CB2R enhances both immunosuppressive TAM infiltration and systemic T-cell suppression through CB2R activation, and that inhibition of CB2Rs can potently counter both the immunosuppressive tumor microenvironment, as well as systemic immunosuppression in glioblastoma.
Collapse
Affiliation(s)
- Jin Duan
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Jieling Chen
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Yilin Lin
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
| | - Stanley L Lin
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, China
| | - Jie Wu
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| |
Collapse
|
23
|
Doron O, Wong T, Ablyazova F, Singha S, Cavallaro J, Ben-Shalom N, D'Amico RS, Harshan M, McKeown A, Zlochower A, Langer DJ, Boockvar JA. Results from a first-in-human phase I safety trial to evaluate the use of a vascularized pericranial/temporoparietal fascial flap to line the resection cavity following resection of newly diagnosed glioblastoma. J Neurooncol 2024; 168:225-235. [PMID: 38664311 PMCID: PMC11147875 DOI: 10.1007/s11060-024-04647-w] [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: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE The efficacy of systemic therapies for glioblastoma (GBM) remains limited due to the constraints of systemic toxicity and blood-brain barrier (BBB) permeability. Temporoparietal fascial flaps (TPFFs) and vascularized peri cranial flaps (PCF) are not restricted by the blood-brain barrier (BBB), as they derive their vascular supply from branches of the external carotid artery. Transposition of a vascularized TPFF or PCF along a GBM resection cavity may bring autologous tissue not restricted by the BBB in close vicinity to the tumor bed microenvironment, permit ingrowth of vascular channels fed by the external circulation, and offer a mechanism of bypassing the BBB. In addition, circulating immune cells in the vascularized flap may have better access to tumor-associated antigens (TAA) within the tumor microenvironment. We conducted a first-in-human Phase I trial assessing the safety of lining the resection cavity with autologous TPFF/PCF of newly diagnosed patients with GBM. METHODS 12 patients underwent safe, maximal surgical resection of newly diagnosed GBMs, followed by lining of the resection cavity with a pedicled, autologous TPFF or PCF. Safety was assessed by monitoring adverse events. Secondary analysis of efficacy was examined as the proportion of patients experiencing progression-free disease (PFS) as indicated by response assessment in neuro-oncology (RANO) criteria and overall survival (OS). The study was powered to determine whether a Phase II study was warranted based on these early results. For this analysis, subjects who were alive and had not progressed as of the date of the last follow-up were considered censored and all living patients who were alive as of the date of last follow-up were considered censored for overall survival. For simplicity, we assumed that a 70% PFS rate at 6 months would be considered an encouraging response and would make an argument for further investigation of the procedure. RESULTS Median age of included patients was 57 years (range 46-69 years). All patients were Isocitrate dehydrogenase (IDH) wildtype. Average tumor volume was 56.6 cm3 (range 14-145 cm3). Resection was qualified as gross total resection (GTR) of all of the enhancing diseases in all patients. Grade III or above adverse events were encountered in 3 patients. No Grade IV or V serious adverse events occurred in the immediate post-operative period including seizure, infection, stroke, or tumor growing along the flap. Disease progression at the site of the original tumor was identified in only 4 (33%) patients (median 23 months, range 8-25 months), 3 of whom underwent re-operation. Histopathological analyses of those implanted flaps and tumor bed biopsy at repeat surgery demonstrated robust immune infiltrates within the transplanted flap. Importantly, no patient demonstrated evidence of tumor infiltration into the implanted flap. At the time of this manuscript preparation, only 4/12 (33%) of patients have died. Based on the statistical considerations above and including all 12 patients 10/12 (83.3%) had 6-month PFS. The median PFS was 9.10 months, and the OS was 17.6 months. 4/12 (33%) of patients have been alive for more than two years and our longest surviving patient currently is alive at 60 months. CONCLUSIONS This pilot study suggests that insertion of pedicled autologous TPFF/PCF along a GBM resection cavity is safe and feasible. Based on the encouraging response rate in 6-month PFS and OS, larger phase II studies are warranted to assess and reproduce safety, feasibility, and efficacy. TRIAL REGISTRATION NUMBER AND DATE OF REGISTRATION FOR PROSPECTIVELY REGISTERED TRIALS: ClinicalTrials.gov ID NCT03630289, dated: 08/02/2018.
Collapse
Affiliation(s)
- Omer Doron
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
- Department of Biomedical Engineering, The Aldar and Iby Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Tamika Wong
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Faina Ablyazova
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Souvik Singha
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Julianna Cavallaro
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Netanel Ben-Shalom
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Randy S D'Amico
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Manju Harshan
- Department of Pathology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Amy McKeown
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Avraham Zlochower
- Department of Radiology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - David J Langer
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - John A Boockvar
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA.
| |
Collapse
|
24
|
Uthanaphun T, Manochantr S, Tantrawatpan C, Tantikanlayaporn D, Kheolamai P. PL-hMSC and CH-hMSC derived soluble factors inhibit proliferation but improve hGBM cell migration by activating TGF-β and inhibiting Wnt signaling. Biosci Rep 2024; 44:BSR20231964. [PMID: 38687607 PMCID: PMC11130542 DOI: 10.1042/bsr20231964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/18/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive brain tumors. GBM resists most chemotherapeutic agents, resulting in a high mortality rate in patients. Human mesenchymal stem cells (hMSCs), which are parts of the cancer stroma, have been shown to be involved in the development and progression of GBM. However, different sources of hMSCs might affect GBM cells differently. In the present study, we established hMSCs from placenta (PL-hMSC) and chorion (CH-hMSC) to study the effects of their released soluble factors on the proliferation, migration, invasion, gene expression, and survival of human GBM cells, U251. We found that the soluble factors derived from CH-hMSCs and PL-hMSCs suppressed the proliferation of U251 cells in a dose-dependent manner. In contrast, soluble factors derived from both hMSC sources increased U251 migration without affecting their invasive property. The soluble factors derived from these hMSCs decreased the expression levels of CyclinD1, E2Fs and MYC genes that promote GBM cell proliferation but increased the expression level of TWIST gene, which promotes EMT and GBM cell migration. The functional study suggests that both hMSCs might exert their effects, at least in part, by activating TGF-β and suppressing Wnt/β-catenin signaling in U251 cells. Our study provides a better understanding of the interaction between GBM cells and gestational tissue-derived hMSCs. This knowledge might be used to develop safer and more effective stem cell therapy that improves the survival and quality of life of patients with GBM by manipulating the interaction between hMSCs and GBM cells.
Collapse
Affiliation(s)
- Tanawat Uthanaphun
- Master of Science Program in Stem Cell and Molecular Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Sirikul Manochantr
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Chairat Tantrawatpan
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pakpoom Kheolamai
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| |
Collapse
|
25
|
Fang Y, Li X, Tian R. Unlocking Glioblastoma Vulnerabilities with CRISPR-Based Genetic Screening. Int J Mol Sci 2024; 25:5702. [PMID: 38891890 PMCID: PMC11171782 DOI: 10.3390/ijms25115702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults. Despite advancements in treatment, the prognosis for patients with GBM remains poor due to its aggressive nature and resistance to therapy. CRISPR-based genetic screening has emerged as a powerful tool for identifying genes crucial for tumor progression and treatment resistance, offering promising targets for tumor therapy. In this review, we provide an overview of the recent advancements in CRISPR-based genetic screening approaches and their applications in GBM. We highlight how these approaches have been used to uncover the genetic determinants of GBM progression and responsiveness to various therapies. Furthermore, we discuss the ongoing challenges and future directions of CRISPR-based screening methods in advancing GBM research.
Collapse
Affiliation(s)
- Yitong Fang
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.F.); (X.L.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xing Li
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.F.); (X.L.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruilin Tian
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.F.); (X.L.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
26
|
Tahmasebi Dehkordi H, Khaledi F, Ghasemi S. Immunological processes of enhancers and suppressors of long non-coding RNAs associated with brain tumors and inflammation. Int Rev Immunol 2024; 43:178-196. [PMID: 37974420 DOI: 10.1080/08830185.2023.2280581] [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: 07/16/2022] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Immunological processes, such as inflammation, can both cause tumor suppression and cancer progression. Moreover, deregulated levels of long non-coding RNA (lncRNA) expression in the brain may cause inflammation and lead to the growth of tumors. Like other biological processes, the immune system's role in cancer is complicated, varies, and can help or hurt the cancer's maintenance. According to research, inflammation and brain cancer are correlated via several signaling pathways. A variety of lncRNAs have recently been revealed to influence cancer by modulating inflammatory pathways. As a result, lncRNAs have the potential to influence carcinogenesis, tumor formation, or tumor suppression via an increase or decrease in inflammation functions. Although the study and targeting of lncRNAs have made great progress in the treatment of cancer, there are definitely limitations and challenges. Using new technologies like nanocarriers and cell-penetrating peptides (CPPs) to target treatments without hurting healthy body tissues has shown to be very effective. In this review article, we have collected significantly related lncRNAs and their inhibitory or stimulating roles in inflammation and brain cancer for the first time. However, there are limitations, such as side effects and damage to normal tissues. With the advancement of new targeting technologies, these lncRNAs may be candidates for the specific targeting therapy of brain cancers by limiting inflammation or stimulating the immune system against them in the future.
Collapse
Affiliation(s)
- Hossein Tahmasebi Dehkordi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Khaledi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
27
|
Deng Y, Chen Q, Wan C, Sun Y, Huang F, Hu Y, Yang K. Microglia and macrophage metabolism: a regulator of cerebral gliomas. Cell Biosci 2024; 14:49. [PMID: 38632627 PMCID: PMC11022384 DOI: 10.1186/s13578-024-01231-7] [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: 12/05/2023] [Accepted: 04/07/2024] [Indexed: 04/19/2024] Open
Abstract
Reciprocal interactions between the tumor microenvironment (TME) and cancer cells play important roles in tumorigenesis and progression of glioma. Glioma-associated macrophages (GAMs), either of peripheral origin or representing brain-intrinsic microglia, are the majority population of infiltrating immune cells in glioma. GAMs, usually classified into M1 and M2 phenotypes, have remarkable plasticity and regulate tumor progression through different metabolic pathways. Recently, research efforts have increasingly focused on GAMs metabolism as potential targets for glioma therapy. This review aims to delineate the metabolic characteristics of GAMs within the TME and provide a summary of current therapeutic strategies targeting GAMs metabolism in glioma. The goal is to provide novel insights and therapeutic pathways for glioma by highlighting the significance of GAMs metabolism.
Collapse
Affiliation(s)
- Yue Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qinyan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
28
|
Thiruppathi J, Vijayan V, Park IK, Lee SE, Rhee JH. Enhancing cancer immunotherapy with photodynamic therapy and nanoparticle: making tumor microenvironment hotter to make immunotherapeutic work better. Front Immunol 2024; 15:1375767. [PMID: 38646546 PMCID: PMC11026591 DOI: 10.3389/fimmu.2024.1375767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Cancer immunotherapy has made tremendous advancements in treating various malignancies. The biggest hurdle to successful immunotherapy would be the immunosuppressive tumor microenvironment (TME) and low immunogenicity of cancer cells. To make immunotherapy successful, the 'cold' TME must be converted to 'hot' immunostimulatory status to activate residual host immune responses. To this end, the immunosuppressive equilibrium in TME should be broken, and immunogenic cancer cell death ought to be induced to stimulate tumor-killing immune cells appropriately. Photodynamic therapy (PDT) is an efficient way of inducing immunogenic cell death (ICD) of cancer cells and disrupting immune-restrictive tumor tissues. PDT would trigger a chain reaction that would make the TME 'hot' and have ICD-induced tumor antigens presented to immune cells. In principle, the strategic combination of PDT and immunotherapy would synergize to enhance therapeutic outcomes in many intractable tumors. Novel technologies employing nanocarriers were developed to deliver photosensitizers and immunotherapeutic to TME efficiently. New-generation nanomedicines have been developed for PDT immunotherapy in recent years, which will accelerate clinical applications.
Collapse
Affiliation(s)
- Jayalakshmi Thiruppathi
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy Medical Research Center (MRC), Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| | - Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - In-Kyu Park
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Radiology, Biomolecular Theranostics (BiT) Laboratory, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy Medical Research Center (MRC), Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| |
Collapse
|
29
|
Wang H, Medina R, Ye J, Zhang Y, Chakraborty S, Valenzuela A, Uher O, Hadrava Vanova K, Sun M, Sang X, Park DM, Zenka J, Gilbert MR, Pacak K, Zhuang Z. rWTC-MBTA Vaccine Induces Potent Adaptive Immune Responses Against Glioblastomas via Dynamic Activation of Dendritic Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308280. [PMID: 38298111 PMCID: PMC11005728 DOI: 10.1002/advs.202308280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Despite strides in immunotherapy, glioblastoma multiforme (GBM) remains challenging due to low inherent immunogenicity and suppressive tumor microenvironment. Converting "cold" GBMs to "hot" is crucial for immune activation and improved outcomes. This study comprehensively characterized a therapeutic vaccination strategy for preclinical GBM models. The vaccine consists of Mannan-BAM-anchored irradiated whole tumor cells, Toll-like receptor ligands [lipoteichoic acid (LTA), polyinosinic-polycytidylic acid (Poly (I:C)), and resiquimod (R-848)], and anti-CD40 agonistic antibody (rWTC-MBTA). Intracranial GBM models (GL261, SB28 cells) are used to evaluate the vaccine efficacy. A substantial number of vaccinated mice exhibited complete regression of GBM tumors in a T-cell-dependent manner, with no significant toxicity. Long-term tumor-specific immune memory is confirmed upon tumor rechallenge. In the vaccine-draining lymph nodes of the SB28 model, rWTC-MBTA vaccination triggered a major rise in conventional dendritic cell type 1 (cDC1) 12 h post-treatment, followed by an increase in conventional dendritic cell type 2 (cDC2), monocyte-derived dendritic cell (moDC), and plasmacytoid dendritic cell (pDC) on Day 5 and Day 13. Enhanced cytotoxicity of CD4+ and CD8+ T cells in vaccinated mice is verified in co-culture with tumor cells. Analyses of immunosuppressive signals (T-cell exhaustion, myeloid-derived suppressor cells (MDSC), M2 macrophages) in the GBM microenvironment suggest potential combinations with other immunotherapies for enhanced efficacy. In conclusion, the authors findings demonstrate that rWTC-MBTA induces potent and long-term adaptive immune responses against GBM.
Collapse
Affiliation(s)
- Herui Wang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Staff Scientist Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100437 Convent Dr.BethesdaMD20892USA
| | - Rogelio Medina
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Juan Ye
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Yaping Zhang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | | | - Alex Valenzuela
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Ondrej Uher
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Katerina Hadrava Vanova
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Mitchell Sun
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Xueyu Sang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Deric M. Park
- John Theurer Cancer CenterHUMCHackensack Meridian School of Medicine92 2nd StHackensackNJ07601USA
| | - Jan Zenka
- Department of Medical BiologyFaculty of ScienceUniversity of South BohemiaČeské Budějovice37005Czech Republic
| | - Mark R. Gilbert
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Zhengping Zhuang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Senior Investigator Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100037 Convent DrBethesdaMD20892USA
| |
Collapse
|
30
|
Cui X, Wang Q, Liu X, Kang C. Levetiracetam: A Potent Sword against Microglia Polarization in Gliomas. Clin Cancer Res 2024; 30:1073-1075. [PMID: 38170191 DOI: 10.1158/1078-0432.ccr-23-3322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
Crosstalk between tumor cells and peritumoral cells contributes to immunosuppressive microenvironment formation in glioblastomas (GBM). A recent study revealed that glioma stem cells activated neuronal activity to promote microglial M2 polarization, leading to GBM progression, which could be pharmacologically blocked by levetiracetam, providing a practical strategy for GBM immunotherapy. See related article by Guo et al., p. 1160.
Collapse
Affiliation(s)
- Xiaoteng Cui
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, P.R China
- Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, P.R. China
| | - Qixue Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, P.R China
- Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, P.R. China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Nankai University, Tianjin, P.R. China
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, P.R China
- Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, P.R. China
| |
Collapse
|
31
|
Qin X, Wang B, Lu X, Song Y, Wang W. Identification and Validation of a PEX5-Dependent Signature for Prognostic Prediction in Glioma. Biomolecules 2024; 14:314. [PMID: 38540734 PMCID: PMC10967733 DOI: 10.3390/biom14030314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Gliomas, the most prevalent and lethal form of brain cancer, are known to exhibit metabolic alterations that facilitate tumor growth, invasion, and resistance to therapies. Peroxisomes, essential organelles responsible for fatty acid oxidation and reactive oxygen species (ROS) homeostasis, rely on the receptor PEX5 for the import of metabolic enzymes into their matrix. However, the prognostic significance of peroxisomal enzymes for glioma patients remains unclear. In this study, we elucidate that PEX5 is indispensable for the cell growth, migration, and invasion of glioma cells. We establish a robust prognosis model based on the expression of peroxisomal enzymes, whose localization relies on PEX5. This PEX5-dependent signature not only serves as a robust prognosis model capable of accurately predicting outcomes for glioma patients, but also effectively distinguishes several clinicopathological features, including the grade, isocitrate dehydrogenase (IDH) mutation, and 1p19q codeletion status. Furthermore, we developed a nomogram that integrates the prognostic model with other clinicopathological factors, demonstrating highly accurate performance in estimating patient survival. Patients classified into the high-risk group based on our prognostic model exhibited an immunosuppressive microenvironment. Finally, our validation reveals that the elevated expression of GSTK1, an antioxidant enzyme within the signature, promotes the cell growth and migration of glioma cells, with this effect dependent on the peroxisomal targeting signal recognized by PEX5. These findings identify the PEX5-dependent signature as a promising prognostic tool for gliomas.
Collapse
Affiliation(s)
| | | | | | | | - Wei Wang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| |
Collapse
|
32
|
Gershon R, Polevikov A, Karepov Y, Shenkar A, Ben-Horin I, Alter Regev T, Dror-Levinsky M, Lipczyc K, Gasri-Plotnitsky L, Diamant G, Shapira N, Bensimhon B, Hagai A, Shahar T, Grossman R, Ram Z, Volovitz I. Frequencies of 4 tumor-infiltrating lymphocytes potently predict survival in glioblastoma, an immune desert. Neuro Oncol 2024; 26:473-487. [PMID: 37870293 PMCID: PMC10912003 DOI: 10.1093/neuonc/noad204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND GBM is an aggressive grade 4 primary brain tumor (BT), with a 5%-13% 5-year survival. Most human GBMs manifest as immunologically "cold" tumors or "immune deserts," yet the promoting or suppressive roles of specific lymphocytes within the GBM tumor microenvironment (TME) is of considerable debate. METHODS We used meticulous multiparametric flow cytometry (FC) to determine the lymphocytic frequencies in 102 GBMs, lower-grade gliomas, brain metastases, and nontumorous brain specimen. FC-attained frequencies were compared with frequencies estimated by "digital cytometry." The FC-derived data were combined with the patients' demographic, clinical, molecular, histopathological, radiological, and survival data. RESULTS Comparison of FC-derived data to CIBERSORT-estimated data revealed the poor capacity of digital cytometry to estimate cell frequencies below 0.2%, the frequency range of most immune cells in BTs. Isocitrate dehydrogenase (IDH) mutation status was found to affect TME composition more than the gliomas' pathological grade. Combining FC and survival data disclosed that unlike other cancer types, the frequency of helper T cells (Th) and cytotoxic T lymphocytes (CTL) correlated negatively with glioma survival. In contrast, the frequencies of γδ-T cells and CD56bright natural killer cells correlated positively with survival. A composite parameter combining the frequencies of these 4 tumoral lymphocytes separated the survival curves of GBM patients with a median difference of 10 months (FC-derived data; P < .0001, discovery cohort), or 4.1 months (CIBERSORT-estimated data; P = .01, validation cohort). CONCLUSIONS The frequencies of 4 TME lymphocytes strongly correlate with the survival of patients with GBM, a tumor considered an immune desert.
Collapse
Affiliation(s)
- Rotem Gershon
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Antonina Polevikov
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Yevgeny Karepov
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Anatoly Shenkar
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Idan Ben-Horin
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Oncology Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Tal Alter Regev
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Meytal Dror-Levinsky
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Kelly Lipczyc
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Lital Gasri-Plotnitsky
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Gil Diamant
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Nati Shapira
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Barak Bensimhon
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Aharon Hagai
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Tal Shahar
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Rachel Grossman
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Zvi Ram
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Ilan Volovitz
- The Cancer Immunotherapy Laboratory, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
- Neurosurgery Department, The Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| |
Collapse
|
33
|
Liao C, Chen W, Xu G, Wang J, Dong W. High expression of ARPC1B correlates with immune infiltration and poor outcomes in glioblastoma. Biochem Biophys Rep 2024; 37:101619. [PMID: 38188361 PMCID: PMC10770598 DOI: 10.1016/j.bbrep.2023.101619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024] Open
Abstract
Objective To investigate the role of ARPC1B in GBM and its prognostic value. Methods mRNA and protein expression of ARPC1B in GBM was analyzed using the TCGA; TIMER2 and the HPA databases, and protein expression differences were detected using immunohistochemistry. K-M analysis and Cox regression analysis were performed on high and low ARPC1B expression groups in the TCGA database. The relationship between immune cells and ARPC1B expression was explored using the TIMER2 database. GO and KEGG analyses were conducted to investigate the functions of ARPC1B-related genes in GBM. Results ARPC1B was highly expressed in both GBM tissues and cell lines, and it was demonstrated as a prognostic biomarker for GBM. ARPC1B expression levels showed associations with immune cell populations within the GBM microenvironment. Conclusion ARPC1B can regulating immune infiltration in the GBM microenvironment, indicating its potential as a novel therapeutic target for GBM.
Collapse
Affiliation(s)
- Chuangxin Liao
- Corresponding author. Department of Neurosurgery, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-sen University, 183 Huangpu Road East, Guangzhou, 510700, China.
| | | | | | - Jingshan Wang
- Department of Neurosurgery, The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijie Dong
- Department of Neurosurgery, The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Mistry AM, Daneshmand J, Seo SJ, Lehman NL, Miller DM, Goodin DA, Frieboes HB, Chen J, Masters A, Williams BJ, Yaddanapudi K. Spatially Resolved Microglia/Macrophages in Recurrent Glioblastomas Overexpress Fatty Acid Metabolism and Phagocytic Genes. Curr Oncol 2024; 31:1183-1194. [PMID: 38534921 PMCID: PMC10968993 DOI: 10.3390/curroncol31030088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) tumors are rich in tumor-associated microglia/macrophages. Changes associated with treatment in this specific cell population are poorly understood. Therefore, we studied changes in gene expression of tumor-associated microglia/macrophages (Iba1+) cells in de novo versus recurrent GBMs. METHODS NanoString GeoMx® Digital Spatial Transcriptomic Profiling of microglia/macrophages (Iba1+) and glial cells (Gfap+) cells identified on tumor sections was performed on paired de novo and recurrent samples obtained from three IDH-wildtype GBM patients. The impact of differentially expressed genes on patient survival was evaluated using publicly available data. RESULTS Unsupervised analyses of the NanoString GeoMx® Digital Spatial Profiling data revealed clustering based on the transcriptomic data from Iba1+ and Gfap+ cells. As expected, conventional differential gene expression and enrichment analyses revealed upregulation of immune-function-related genes in Iba1+ cells compared to Gfap+ cells. A focused differential gene expression analysis revealed upregulation of phagocytosis and fatty acid/lipid metabolism genes in Iba1+ cells in recurrent GBM samples compared to de novo GBM samples. Importantly, of these genes, the lipid metabolism gene PLD3 consistently correlated with survival in multiple different publicly available datasets. CONCLUSION Tumor-associated microglia/macrophages in recurrent GBM overexpress genes involved in fatty acid/lipid metabolism. Further investigation is needed to fully delineate the role of PLD phospholipases in GBM progression.
Collapse
Affiliation(s)
- Akshitkumar M. Mistry
- Department of Neurosurgery, University of Louisville, Louisville, KY 40202, USA; (S.J.S.); (B.J.W.)
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; (D.M.M.); (H.B.F.); (A.M.)
| | - Jonah Daneshmand
- Department of Bioinformatics, University of Louisville, Louisville, KY 40202, USA;
| | - SeonYeong Jamie Seo
- Department of Neurosurgery, University of Louisville, Louisville, KY 40202, USA; (S.J.S.); (B.J.W.)
| | - Norman L. Lehman
- Departments of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Donald M. Miller
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; (D.M.M.); (H.B.F.); (A.M.)
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Dylan A. Goodin
- Department of Bioengineering, University of Louisville, Louisville, KY 40202, USA; (D.A.G.); (J.C.)
| | - Hermann B. Frieboes
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; (D.M.M.); (H.B.F.); (A.M.)
- Department of Bioengineering, University of Louisville, Louisville, KY 40202, USA; (D.A.G.); (J.C.)
| | - Joseph Chen
- Department of Bioengineering, University of Louisville, Louisville, KY 40202, USA; (D.A.G.); (J.C.)
| | - Adrianna Masters
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; (D.M.M.); (H.B.F.); (A.M.)
- Department of Radiation Oncology, University of Louisville, Louisville, KY 40202, USA
| | - Brian J. Williams
- Department of Neurosurgery, University of Louisville, Louisville, KY 40202, USA; (S.J.S.); (B.J.W.)
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; (D.M.M.); (H.B.F.); (A.M.)
| | - Kavitha Yaddanapudi
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; (D.M.M.); (H.B.F.); (A.M.)
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
- Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
35
|
Lootens T, Roman BI, Stevens CV, De Wever O, Raedt R. Glioblastoma-Associated Mesenchymal Stem/Stromal Cells and Cancer-Associated Fibroblasts: Partners in Crime? Int J Mol Sci 2024; 25:2285. [PMID: 38396962 PMCID: PMC10889514 DOI: 10.3390/ijms25042285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated mesenchymal stem/stromal cells (TA-MSCs) have been recognized as attractive therapeutic targets in several cancer types, due to their ability to enhance tumor growth and angiogenesis and their contribution to an immunosuppressive tumor microenvironment (TME). In glioblastoma (GB), mesenchymal stem cells (MSCs) seem to be recruited to the tumor site, where they differentiate into glioblastoma-associated mesenchymal stem/stromal cells (GA-MSCs) under the influence of tumor cells and the TME. GA-MSCs are reported to exert important protumoral functions, such as promoting tumor growth and invasion, increasing angiogenesis, stimulating glioblastoma stem cell (GSC) proliferation and stemness, mediating resistance to therapy and contributing to an immunosuppressive TME. Moreover, they could act as precursor cells for cancer-associated fibroblasts (CAFs), which have recently been identified in GB. In this review, we provide an overview of the different functions exerted by GA-MSCs and CAFs and the current knowledge on the relationship between these cell types. Increasing our understanding of the interactions and signaling pathways in relevant models might contribute to future regimens targeting GA-MSCs and GB-associated CAFs to inhibit tumor growth and render the TME less immunosuppressive.
Collapse
Affiliation(s)
- Thibault Lootens
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium;
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
| | - Bart I. Roman
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
- SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Christian V. Stevens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
- SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
| | - Robrecht Raedt
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; (B.I.R.); (C.V.S.)
| |
Collapse
|
36
|
Romagnoli G, D'Alessandris QG, Capone I, Tavilla A, Canini I, Lapenta C, Buccarelli M, Giordano M, Tirelli V, Sanchez M, Fragale A, Giannetti S, Di Bonaventura R, Lauretti L, Biffoni M, Ricci-Vitiani L, Pallini R, Gabriele L. CD8+CD103+PD1+TIM3+ T cells in glioblastoma microenvironment correlate with prognosis. Immunology 2024; 171:198-211. [PMID: 37884280 DOI: 10.1111/imm.13710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Glioblastoma, isocitrate dehydrogenase-wildtype (GB), is the most common and aggressive primary brain malignancy with poor outcome. Immune checkpoint inhibitors (ICIs) have been tested in GB and, despite disappointing results, the identification of a small subgroup of responders underlies the need to improve our understanding of the tumour microenvironment (TME) immunity. This study aimed to determine whether the expression of selected immune checkpoints on tissue-resident memory T cells (Trm) may predict patient outcome. We conducted a single cohort observational study. Tumour samples were collected from 45 patients with histologically confirmed GB (WHO grade 4) and processed to obtain single-cell suspensions. Patients were assessed for the correlation of Trm phenotype with overall survival (OS) or progression-free survival (PFS) using multiparametric flow cytometry and uni/multivariate analyses. Levels of Trm expressing programmed cell death protein 1 (PD1) and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) were found to be linked to clinical outcome. Low frequency of Trm expressing PD1 or TIM3 or both markers defined subgroups as independent positive prognostic factors for patient survival. On multivariate analysis, low CD8+CD103+PD1+TIM3+ Trm and Karnofsky performance status (KPS) ≥70 were confirmed to be the most predictive independent factors associated with longer OS (hazard ratios-HR [95%CI]: 0.14 [0.04-0.52] p < 0.001, 0.39 [0.16-0.96] p = 0.04, respectively). The CD8+CD103+ Trm subgroups were also age-related predictors for survival in GB.
Collapse
Affiliation(s)
- Giulia Romagnoli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Quintino Giorgio D'Alessandris
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Imerio Capone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Tavilla
- National Centre for Disease Prevention and Health Promotion, Istituto Superiore di Sanità, Rome, Italy
| | - Irene Canini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Caterina Lapenta
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Giordano
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Alessandra Fragale
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Giannetti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rina Di Bonaventura
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Liverana Lauretti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Pallini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
37
|
Losurdo A, Di Muzio A, Cianciotti BC, Dipasquale A, Persico P, Barigazzi C, Bono B, Feno S, Pessina F, Santoro A, Simonelli M. T Cell Features in Glioblastoma May Guide Therapeutic Strategies to Overcome Microenvironment Immunosuppression. Cancers (Basel) 2024; 16:603. [PMID: 38339353 PMCID: PMC10854506 DOI: 10.3390/cancers16030603] [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: 12/28/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive and lethal primary brain tumor, bearing a survival estimate below 10% at five years, despite standard chemoradiation treatment. At recurrence, systemic treatment options are limited and the standard of care is not well defined, with inclusion in clinical trials being highly encouraged. So far, the use of immunotherapeutic strategies in GBM has not proved to significantly improve patients' prognosis in the treatment of newly diagnosed GBM, nor in the recurrent setting. Probably this has to do with the unique immune environment of the central nervous system, which harbors several immunosuppressive/pro-tumorigenic factors, both soluble (e.g., TGF-β, IL-10, STAT3, prostaglandin E2, and VEGF) and cellular (e.g., Tregs, M2 phenotype TAMs, and MDSC). Here we review the immune composition of the GBMs microenvironment, specifically focusing on the phenotype and function of the T cell compartment. Moreover, we give hints on the therapeutic strategies, such as immune checkpoint blockade, vaccinations, and adoptive cell therapy, that, interacting with tumor-infiltrating lymphocytes, might both target in different ways the tumor microenvironment and potentiate the activity of standard therapies. The path to be followed in advancing clinical research on immunotherapy for GBM treatment relies on a twofold strategy: testing combinatorial treatments, aiming to restore active immune anti-tumor responses, tackling immunosuppression, and additionally, designing more phase 0 and window opportunity trials with solid translational analyses to gain deeper insight into the on-treatment shaping of the GBM microenvironment.
Collapse
Affiliation(s)
- Agnese Losurdo
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Antonio Di Muzio
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Beatrice Claudia Cianciotti
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (B.C.C.); (S.F.)
| | - Angelo Dipasquale
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
| | - Pasquale Persico
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Chiara Barigazzi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Beatrice Bono
- Department of Neurosurgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Simona Feno
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (B.C.C.); (S.F.)
| | - Federico Pessina
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- Department of Neurosurgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Armando Santoro
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Matteo Simonelli
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (A.L.); (A.D.M.); (A.D.); (P.P.); (C.B.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| |
Collapse
|
38
|
Cui X, Huo D, Wang Q, Wang Y, Liu X, Zhao K, You Y, Zhang J, Kang C. RUNX1/NPM1/H3K4me3 complex contributes to extracellular matrix remodeling via enhancing FOSL2 transcriptional activation in glioblastoma. Cell Death Dis 2024; 15:98. [PMID: 38286983 PMCID: PMC10825180 DOI: 10.1038/s41419-024-06481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 01/31/2024]
Abstract
Extracellular matrix (ECM) remodeling has been implicated in the tumor malignant progression and immune escape in glioblastoma (GBM). Runt-related transcription factor 1 (RUNX1) is a vital transcriptional factor for promoting tumorigenesis and invasion in mesenchymal subtype of GBM. But the correlation between RUNX1 and ECM genes expression and regulatory mechanism of RUNX1 on ECM genes expression remain poorly understood to date. In this study, by using integral analysis of chromatin immunoprecipitation-sequencing and RNA sequencing, we reported that RUNX1 positively regulated the expression of various ECM-related genes, including Fibronectin 1 (FN1), Collagen type IV alpha 1 chain (COL4A1), and Lumican (LUM), in GBM. Mechanistically, we demonstrated that RUNX1 interacted with Nucleophosmin 1 (NPM1) to maintain the chromatin accessibility and facilitate FOS Like 2, AP-1 Transcription Factor Subunit (FOSL2)-mediated transcriptional activation of ECM-related genes, which was independent of RUNX1's transcriptional function. ECM remodeling driven by RUNX1 promoted immunosuppressive microenvironment in GBM. In conclusion, this study provides a novel mechanism of RUNX1 binding to NPM1 in driving the ECM remodeling and GBM progression.
Collapse
Affiliation(s)
- Xiaoteng Cui
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Dawei Huo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Laboratory, Institute of Hematology, Zhejiang University, Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Qixue Wang
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Yunfei Wang
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Nankai University, Tianjin, 300350, China
| | - Kai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China.
| | - Chunsheng Kang
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China.
| |
Collapse
|
39
|
Boyineni J, Wood JM, Ravindra A, Boley E, Donohue SE, Soares MB, Malchenko S. Prospective Approach to Deciphering the Impact of Intercellular Mitochondrial Transfer from Human Neural Stem Cells and Brain Tumor-Initiating Cells to Neighboring Astrocytes. Cells 2024; 13:204. [PMID: 38334595 PMCID: PMC10854889 DOI: 10.3390/cells13030204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/10/2024] Open
Abstract
The communication between neural stem cells (NSCs) and surrounding astrocytes is essential for the homeostasis of the NSC niche. Intercellular mitochondrial transfer, a unique communication system that utilizes the formation of tunneling nanotubes for targeted mitochondrial transfer between donor and recipient cells, has recently been identified in a wide range of cell types. Intercellular mitochondrial transfer has also been observed between different types of cancer stem cells (CSCs) and their neighboring cells, including brain CSCs and astrocytes. CSC mitochondrial transfer significantly enhances overall tumor progression by reprogramming neighboring cells. Despite the urgent need to investigate this newly identified phenomenon, mitochondrial transfer in the central nervous system remains largely uncharacterized. In this study, we found evidence of intercellular mitochondrial transfer from human NSCs and from brain CSCs, also known as brain tumor-initiating cells (BTICs), to astrocytes in co-culture experiments. Both NSC and BTIC mitochondria triggered similar transcriptome changes upon transplantation into the recipient astrocytes. In contrast to NSCs, the transplanted mitochondria from BTICs had a significant proliferative effect on the recipient astrocytes. This study forms the basis for mechanistically deciphering the impact of intercellular mitochondrial transfer on recipient astrocytes, which will potentially provide us with new insights into the mechanisms of mitochondrial retrograde signaling.
Collapse
Affiliation(s)
- Jerusha Boyineni
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| | - Jason Michael Wood
- Research Informatics Core, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Aditya Ravindra
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| | - Ethan Boley
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| | - Sarah E. Donohue
- Research Services, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA;
| | - Marcelo Bento Soares
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
- Department of Psychiatry & Behavioral Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Sergey Malchenko
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (J.B.); (A.R.); (E.B.); (M.B.S.)
| |
Collapse
|
40
|
Chia TY, Billingham LK, Boland L, Katz JL, Arrieta VA, Shireman J, Rosas AL, DeLay SL, Zillinger K, Geng Y, Kruger J, Silvers C, Wang H, Vazquez Cervantes GI, Hou D, Wang S, Wan H, Sonabend A, Zhang P, Lee-Chang C, Miska J. The CXCL16-CXCR6 axis in glioblastoma modulates T-cell activity in a spatiotemporal context. Front Immunol 2024; 14:1331287. [PMID: 38299146 PMCID: PMC10827847 DOI: 10.3389/fimmu.2023.1331287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Glioblastoma multiforme (GBM) pathobiology is characterized by its significant induction of immunosuppression within the tumor microenvironment, predominantly mediated by immunosuppressive tumor-associated myeloid cells (TAMCs). Myeloid cells play a pivotal role in shaping the GBM microenvironment and influencing immune responses, with direct interactions with effector immune cells critically impacting these processes. Methods Our study investigates the role of the CXCR6/CXCL16 axis in T-cell myeloid interactions within GBM tissues. We examined the surface expression of CXCL16, revealing its limitation to TAMCs, while microglia release CXCL16 as a cytokine. The study explores how these distinct expression patterns affect T-cell engagement, focusing on the consequences for T-cell function within the tumor environment. Additionally, we assessed the significance of CXCR6 expression in T-cell activation and the initial migration to tumor tissues. Results Our data demonstrates that CXCL16 surface expression on TAMCs results in predominant T-cell engagement with these cells, leading to impaired T-cell function within the tumor environment. Conversely, our findings highlight the essential role of CXCR6 expression in facilitating T-cell activation and initial migration to tumor tissues. The CXCL16-CXCR6 axis exhibits dualistic characteristics, facilitating the early stages of the T-cell immune response and promoting T-cell infiltration into tumors. However, once inside the tumor, this axis contributes to immunosuppression. Discussion The dual nature of the CXCL16-CXCR6 axis underscores its potential as a therapeutic target in GBM. However, our results emphasize the importance of carefully considering the timing and context of intervention. While targeting this axis holds promise in combating GBM, the complex interplay between TAMCs, microglia, and T cells suggests that intervention strategies need to be tailored to optimize the balance between promoting antitumor immunity and preventing immunosuppression within the dynamic tumor microenvironment.
Collapse
Affiliation(s)
- Tzu-Yi Chia
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Leah K. Billingham
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lauren Boland
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, United States
| | - Joshua L. Katz
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Victor A. Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jack Shireman
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI, United States
| | - Aurora-Lopez Rosas
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Susan L. DeLay
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kaylee Zillinger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Yuheng Geng
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jeandre Kruger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Caylee Silvers
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hanxiang Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gustavo Ignacio Vazquez Cervantes
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - David Hou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Si Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hanxiao Wan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Adam Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Peng Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
41
|
Low JT, Brown MC, Reitman ZJ, Bernstock JD, Markert JM, Friedman GK, Waitkus MS, Bowie ML, Ashley DM. Understanding and therapeutically exploiting cGAS/STING signaling in glioblastoma. J Clin Invest 2024; 134:e163452. [PMID: 38226619 PMCID: PMC10786687 DOI: 10.1172/jci163452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
Since the discovery that cGAS/STING recognizes endogenous DNA released from dying cancer cells and induces type I interferon and antitumor T cell responses, efforts to understand and therapeutically target the STING pathway in cancer have ensued. Relative to other cancer types, the glioma immune microenvironment harbors few infiltrating T cells, but abundant tumor-associated myeloid cells, possibly explaining disappointing responses to immune checkpoint blockade therapies in cohorts of patients with glioblastoma. Notably, unlike most extracranial tumors, STING expression is absent in the malignant compartment of gliomas, likely due to methylation of the STING promoter. Nonetheless, several preclinical studies suggest that inducing cGAS/STING signaling in the glioma immune microenvironment could be therapeutically beneficial, and cGAS/STING signaling has been shown to mediate inflammatory and antitumor effects of other modalities either in use or being developed for glioblastoma therapy, including radiation, tumor-treating fields, and oncolytic virotherapy. In this Review, we discuss cGAS/STING signaling in gliomas, its implications for glioma immunobiology, compartment-specific roles for STING signaling in influencing immune surveillance, and efforts to target STING signaling - either directly or indirectly - for antiglioma therapy.
Collapse
Affiliation(s)
| | | | - Zachary J. Reitman
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - James M. Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gregory K. Friedman
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | |
Collapse
|
42
|
Li J, Song Z, Chen Z, Gu J, Cai Y, Zhang L, Wang Z. Association Between Diverse Cell Death Patterns Related Gene Signature and Prognosis, Drug Sensitivity, and Immune Microenvironment in Glioblastoma. J Mol Neurosci 2024; 74:10. [PMID: 38214842 PMCID: PMC10787010 DOI: 10.1007/s12031-023-02181-4] [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: 08/18/2023] [Accepted: 11/07/2023] [Indexed: 01/13/2024]
Abstract
Glioblastoma (GBM) is the most invasive type of glioma and is difficult to treat. Diverse programmed cell death (PCD) patterns have a significant association with tumor initiation and progression. A novel prognostic model based on PCD genes may serve as an effective tool to predict the prognosis of GBM. The study incorporated 11 PCD patterns, namely apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, entotic cell death, netotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis, and oxeiptosis, to develop the model. To construct and validate the model, both bulk and single-cell transcriptome data, along with corresponding clinical data from GBM cases, were obtained from the TCGA-GBM, REMBRANDT, CGGA, and GSE162631 datasets. A cell death-related signature containing 14 genes was constructed with the TCGA-GBM cohort and validated in the REMBRANDT and CGGA datasets. GBM patients with a higher cell death index (CDI) were significantly associated with poorer survival outcomes. Two separate clusters associated with clinical outcomes emerged from unsupervised analysis. A multivariate Cox regression analysis was conducted to examine the association of CDI with clinical characteristics, and a prognostic nomogram was developed. Drug sensitivity analysis revealed high-CDI GBM patients might be resistant to carmustine while sensitive to 5-fluorouracil. Less abundance of natural killer cells was found in GBM cases with high CDI and bulk transcriptome data. A cell death-related prognostic model that could predict the prognosis of GBM patients with good performance was established, which could discriminate between the prognosis and drug sensitivity of GBM.
Collapse
Affiliation(s)
- Jian Li
- Department of Neurosurgery, Zhangjiagang Hospital affiliated to Soochow University/ The First Peoples' Hospital of Zhangjiagang City, Suzhou, China
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhaoming Song
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhouqing Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jingyu Gu
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yifan Cai
- Department of Neurosurgery, Zhangjiagang Hospital affiliated to Soochow University/ The First Peoples' Hospital of Zhangjiagang City, Suzhou, China
| | - Li Zhang
- Department of Neurosurgery, Zhangjiagang Hospital affiliated to Soochow University/ The First Peoples' Hospital of Zhangjiagang City, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
43
|
Mishchenko TA, Turubanova VD, Gorshkova EN, Krysko O, Vedunova MV, Krysko DV. Glioma: bridging the tumor microenvironment, patient immune profiles and novel personalized immunotherapy. Front Immunol 2024; 14:1299064. [PMID: 38274827 PMCID: PMC10809268 DOI: 10.3389/fimmu.2023.1299064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
Glioma is the most common primary brain tumor, characterized by a consistently high patient mortality rate and a dismal prognosis affecting both survival and quality of life. Substantial evidence underscores the vital role of the immune system in eradicating tumors effectively and preventing metastasis, underscoring the importance of cancer immunotherapy which could potentially address the challenges in glioma therapy. Although glioma immunotherapies have shown promise in preclinical and early-phase clinical trials, they face specific limitations and challenges that have hindered their success in further phase III trials. Resistance to therapy has been a major challenge across many experimental approaches, and as of now, no immunotherapies have been approved. In addition, there are several other limitations facing glioma immunotherapy in clinical trials, such as high intra- and inter-tumoral heterogeneity, an inherently immunosuppressive microenvironment, the unique tissue-specific interactions between the central nervous system and the peripheral immune system, the existence of the blood-brain barrier, which is a physical barrier to drug delivery, and the immunosuppressive effects of standard therapy. Therefore, in this review, we delve into several challenges that need to be addressed to achieve boosted immunotherapy against gliomas. First, we discuss the hurdles posed by the glioma microenvironment, particularly its primary cellular inhabitants, in particular tumor-associated microglia and macrophages (TAMs), and myeloid cells, which represent a significant barrier to effective immunotherapy. Here we emphasize the impact of inducing immunogenic cell death (ICD) on the migration of Th17 cells into the tumor microenvironment, converting it into an immunologically "hot" environment and enhancing the effectiveness of ongoing immunotherapy. Next, we address the challenge associated with the accurate identification and characterization of the primary immune profiles of gliomas, and their implications for patient prognosis, which can facilitate the selection of personalized treatment regimens and predict the patient's response to immunotherapy. Finally, we explore a prospective approach to developing highly personalized vaccination strategies against gliomas, based on the search for patient-specific neoantigens. All the pertinent challenges discussed in this review will serve as a compass for future developments in immunotherapeutic strategies against gliomas, paving the way for upcoming preclinical and clinical research endeavors.
Collapse
Affiliation(s)
- Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victoria D. Turubanova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Neuroscience Research Institute, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ekaterina N. Gorshkova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Dmitri V. Krysko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
44
|
Doherty C, Wilbanks B, Khatua S, Maher LJ. Aptamers in neuro-oncology: An emerging therapeutic modality. Neuro Oncol 2024; 26:38-54. [PMID: 37619244 PMCID: PMC10768989 DOI: 10.1093/neuonc/noad156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Indexed: 08/26/2023] Open
Abstract
Despite recent advances in the understanding of brain tumor pathophysiology, challenges associated with tumor location and characteristics have prevented significant improvement in neuro-oncology therapies. Aptamers are short, single-stranded DNA or RNA oligonucleotides that fold into sequence-specific, 3-dimensional shapes that, like protein antibodies, interact with targeted ligands with high affinity and specificity. Aptamer technology has recently been applied to neuro-oncology as a potential approach to innovative therapy. Preclinical research has demonstrated the ability of aptamers to overcome some obstacles that have traditionally rendered neuro-oncology therapies ineffective. Potential aptamer advantages include their small size, ability in some cases to penetrate the blood-brain barrier, inherent lack of immunogenicity, and applicability for discovering novel biomarkers. Herein, we review recent reports of aptamer applications in neuro-oncology including aptamers found by cell- and in vivo- Systematic Evolution of Ligands by Exponential Enrichment approaches, aptamer-targeted therapeutic delivery modalities, and aptamers in diagnostics and imaging. We further identify crucial future directions for the field that will be important to advance aptamer-based drugs or tools to clinical application in neuro-oncology.
Collapse
Affiliation(s)
- Caroline Doherty
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Medical Scientist Training Program, Mayo Clinic Graduate School of Biomedical Sciences and Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Brandon Wilbanks
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Biochemistry and Molecular Biology Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Section of Neuro-Oncology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Louis James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|
45
|
Nóbrega AHL, Pimentel RS, Prado AP, Garcia J, Frozza RL, Bernardi A. Neuroinflammation in Glioblastoma: The Role of the Microenvironment in Tumour Progression. Curr Cancer Drug Targets 2024; 24:579-594. [PMID: 38310461 DOI: 10.2174/0115680096265849231031101449] [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: 06/30/2023] [Revised: 08/25/2023] [Accepted: 09/08/2023] [Indexed: 02/05/2024]
Abstract
Glioblastoma (GBM) stands as the most aggressive and lethal among the main types of primary brain tumors. It exhibits malignant growth, infiltrating the brain tissue, and displaying resistance toward treatment. GBM is a complex disease characterized by high degrees of heterogeneity. During tumour growth, microglia and astrocytes, among other cells, infiltrate the tumour microenvironment and contribute extensively to gliomagenesis. Tumour-associated macrophages (TAMs), either of peripheral origin or representing brain-intrinsic microglia, are the most numerous nonneoplastic populations in the tumour microenvironment in GBM. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumour microenvironment, which mostly induces tumour aggressiveness and drug resistance. The immunosuppressive tumour microenvironment of GBM provides multiple pathways for tumour immune evasion, contributing to tumour progression. Additionally, TAMs and astrocytes can contribute to tumour progression through the release of cytokines and activation of signalling pathways. In this review, we summarize the role of the microenvironment in GBM progression, focusing on neuroinflammation. These recent advancements in research of the microenvironment hold the potential to offer a promising approach to the treatment of GBM in the coming times.
Collapse
Affiliation(s)
| | - Rafael Sampaio Pimentel
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Ana Paula Prado
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Jenifer Garcia
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Rudimar Luiz Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| | - Andressa Bernardi
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro/RJ, Brazil
| |
Collapse
|
46
|
Yan X, Chen X, Shan Z, Bi L. Engineering Exosomes to Specifically Target the Mitochondria of Brain Cells. ACS OMEGA 2023; 8:48984-48993. [PMID: 38162779 PMCID: PMC10753542 DOI: 10.1021/acsomega.3c06617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
Mitochondrial dysfunction is associated with various health conditions, including cardiovascular and neurodegenerative diseases. Mitochondrial-targeting therapy aims to restore or enhance mitochondrial function to treat or alleviate these conditions. Exosomes, small vesicles that cells secrete, containing a variety of biomolecules, are critical in cell-to-cell communication and have been studied as potential therapeutic agents. Exosome-based therapy has the potential to treat both cardiovascular and neurodegenerative diseases. Combining these two approaches involves using exosomes as carriers to transport mitochondrial-targeting agents to dysfunctional or damaged mitochondria within target cells. This article presents a new technique for engineering brain-derived exosomes that target mitochondria and has demonstrated promise in initial tests with primary neuron cells and healthy rats. This promising development represents a significant step forward in treating these debilitating conditions.
Collapse
Affiliation(s)
- Xin Yan
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health
Research Institute, Michigan Technological
University, Houghton, Michigan 49931, United States
| | - Xinqian Chen
- Department
of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, Michigan 49931, United States
- Health
Research Institute, Michigan Technological
University, Houghton, Michigan 49931, United States
| | - Zhiying Shan
- Department
of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, Michigan 49931, United States
- Health
Research Institute, Michigan Technological
University, Houghton, Michigan 49931, United States
| | - Lanrong Bi
- Department
of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health
Research Institute, Michigan Technological
University, Houghton, Michigan 49931, United States
| |
Collapse
|
47
|
Deng L, Ren J, Li B, Wang Y, Jiang N, Wang Y, Cui H. Predictive value of CCL2 in the prognosis and immunotherapy response of glioblastoma multiforme. BMC Genomics 2023; 24:746. [PMID: 38057698 DOI: 10.1186/s12864-023-09674-x] [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: 03/03/2023] [Accepted: 09/12/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and lethal primary brain tumor with a poor prognosis. The C-C motif chemokine ligand 2 (CCL2) has shown abnormal expression associated with progression of multiple malignancies, however, its role in predicting the prognosis and immunotherapy response of GBM remains poorly understood. RESULTS CCL2 was highly expressed in GBM as analyzed by integrating CGGA, GEPIA and UALCAN online platforms, and further verified by histologic examinations, qRT-PCR analysis, and independent GEO datasets. CCL2 could serve as an independent prognostic factor for both the poor overall survival and progression-free survival of GBM patients based on TCGA data, univariate and multivariate cox analyses. Functional enrichment analysis revealed that CCL2 mainly participated in the regulation of chemokine signaling pathway and inflammatory response. Further, CCL2 expression was positively correlated with CD4 T cells, macrophages, neutrophils and myeloid dendritic cells infiltrating GBM as calculated by the TIMER2.0 algorithm. Importantly, the tumor immune dysfunction and exclusion (TIDE) algorithm showed that in CCL2-high GBM group, the expression of CD274, CTLA4, HAVCR2 and other immune checkpoints were significantly increased, and the immune checkpoint blockade (ICB) therapy was accordingly more responsive. CONCLUSIONS CCL2 can be used as a predictor of prognosis as well as immunotherapy response in GBM, offering potential clinical implications.
Collapse
Affiliation(s)
- Longfei Deng
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Jie Ren
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Benqin Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Yinggang Wang
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Nianfen Jiang
- Health Management Center, Southwest University Hospital, Chongqing, 400715, China
| | - Yi Wang
- Department of Endocrinology, The Ninth People's Hospital of Chongqing, Chongqing, 400799, China.
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400715, China.
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
48
|
Liu W, Zhao Y, Liu Z, Zhang G, Wu H, Zheng X, Tang X, Chen Z. Therapeutic effects against high-grade glioblastoma mediated by engineered induced neural stem cells combined with GD2-specific CAR-NK. Cell Oncol (Dordr) 2023; 46:1747-1762. [PMID: 37420122 DOI: 10.1007/s13402-023-00842-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 07/09/2023] Open
Abstract
PURPOSE High-grade glioblastoma is extremely challenging to treat because of its aggressiveness and resistance to conventional chemo- and radio-therapies. On the contrary, genetic and cellular immunotherapeutic strategies based on the stem and immune cells are emerging as promising treatments against glioblastoma (GBM). We aimed to developed a novel combined immunotherapeutic strategy to improve the treatment efficacy using genetically engineered PBMC-derived induced neural stem cells (iNSCs) expressing HSV-TK and second-generation CAR-NK cells against GBM. METHODS iNSCs cells expressing HSV-TK (iNSCsTK) and GD2-specific CAR-NK92 (GD2NK92) were generated from PBMC-derived iNSCs and NK92 cell lines, respectively. The anti-tumor effect of iNSCsTK and the combinational therapeutics of iNSCsTK and GD2NK92 were evaluated by GBM cell line using in vitro and in vivo experiments. RESULTS PBMC-derived iNSCsTK possessed tumor-tropism migration ability in vitro and in vivo, which exhibited considerable anti-tumor activity via bystander effect in the presence of ganciclovir (GCV). iNSCsTK/GCV could slow GBM progression and prolong median survival in tumor-bearing mice. However, the anti-tumor effect was limited to single therapy. Therefore, the combinational therapeutic effect of iNSCsTK/GCV and GD2NK92 against GBM was investigated. This approach displayed a more significant anti-tumor effect in vitro and in xenograft tumor mice. CONCLUSIONS PBMC-derived iNSCsTK showed a significant tumor-tropic migration and an effective anti-tumor activity with GCV in vitro and in vivo. In addition, combined with GD2NK92, iNSCsTK therapeutic efficacy improved dramatically to prolong the tumor-bearing animal model's median survival.
Collapse
Affiliation(s)
- Weihua Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Guangji Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Xin Zheng
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Xihe Tang
- Neurosurgery Center of Aeronautical General Hospital, Beijing, 100012, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China.
- , Beijing, China.
| |
Collapse
|
49
|
Haydar D, Ibañez-Vega J, Crawford JC, Chou CH, Guy CS, Meehl M, Yi Z, Perry S, Laxton J, Cunningham T, Langfitt D, Vogel P, DeRenzo C, Gottschalk S, Roussel MF, Thomas PG, Krenciute G. CAR T-cell Design-dependent Remodeling of the Brain Tumor Immune Microenvironment Modulates Tumor-associated Macrophages and Anti-glioma Activity. CANCER RESEARCH COMMUNICATIONS 2023; 3:2430-2446. [PMID: 37971169 PMCID: PMC10689147 DOI: 10.1158/2767-9764.crc-23-0424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Understanding the intricate dynamics between adoptively transferred immune cells and the brain tumor immune microenvironment (TIME) is crucial for the development of effective T cell-based immunotherapies. In this study, we investigated the influence of the TIME and chimeric antigen receptor (CAR) design on the anti-glioma activity of B7-H3-specific CAR T-cells. Using an immunocompetent glioma model, we evaluated a panel of seven fully murine B7-H3 CARs with variations in transmembrane, costimulatory, and activation domains. We then investigated changes in the TIME following CAR T-cell therapy using high-dimensional flow cytometry and single-cell RNA sequencing. Our results show that five out of six B7-H3 CARs with single costimulatory domains demonstrated robust functionality in vitro. However, these CARs had significantly varied levels of antitumor activity in vivo. To enhance therapeutic effectiveness and persistence, we incorporated 41BB and CD28 costimulation through transgenic expression of 41BBL on CD28-based CAR T-cells. This CAR design was associated with significantly improved anti-glioma efficacy in vitro but did not result in similar improvements in vivo. Analysis of the TIME revealed that CAR T-cell therapy influenced the composition of the TIME, with the recruitment and activation of distinct macrophage and endogenous T-cell subsets crucial for successful antitumor responses. Indeed, complete brain macrophage depletion using a CSF1R inhibitor abrogated CAR T-cell antitumor activity. In sum, our study highlights the critical role of CAR design and its modulation of the TIME in mediating the efficacy of adoptive immunotherapy for high-grade glioma. SIGNIFICANCE CAR T-cell immunotherapies hold great potential for treating brain cancers; however, they are hindered by a challenging immune environment that dampens their effectiveness. In this study, we show that the CAR design influences the makeup of the immune environment in brain tumors, underscoring the need to target specific immune components to improve CAR T-cell performance, and highlighting the significance of using models with functional immune systems to optimize this therapy.
Collapse
Affiliation(s)
- Dalia Haydar
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
- Children's National Hospital, Center for Cancer and Immunology Research, Washington, District of Columbia
| | - Jorge Ibañez-Vega
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
| | | | - Ching-Heng Chou
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Clifford S. Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michaela Meehl
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
- Department of Microbiology Immunology Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Zhongzhen Yi
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
- Children's National Hospital, Center for Cancer and Immunology Research, Washington, District of Columbia
| | - Scott Perry
- Flow Cytometry Core, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jonathan Laxton
- Flow Cytometry Core, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Trevor Cunningham
- Flow Cytometry Core, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Deanna Langfitt
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Christopher DeRenzo
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
| | - Stephen Gottschalk
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
| | - Martine F. Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Giedre Krenciute
- St. Jude Children's Research Hospital, Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tennessee
| |
Collapse
|
50
|
Wang C, He Z. Integrating bulk and single-cell RNA sequencing data reveals epithelial-mesenchymal transition molecular subtype and signature to predict prognosis, immunotherapy efficacy, and drug candidates in low-grade gliomas. Front Pharmacol 2023; 14:1276466. [PMID: 38053842 PMCID: PMC10694300 DOI: 10.3389/fphar.2023.1276466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Objective: Epithelial-mesenchymal transition (EMT) is a tightly regulated and dynamic process occurring in both embryonic development and tumor progression. Our study aimed to comprehensively explore the molecular subtypes, immune landscape, and prognostic signature based on EMT-related genes in low-grade gliomas (LGG) in order to facilitate treatment decision-making and drug discovery. Methods: We curated EMT-related genes and performed molecular subtyping with consensus clustering algorithm to determine EMT expression patterns in LGG. The infiltration level of diverse immune cell subsets was evaluated by implementing the single-sample gene set enrichment analysis (ssGSEA) and ESTIMATE algorithms. The distinctions in clinical characteristics, mutation landscape, and immune tumor microenvironment (TME) among the subtypes were subjected to further investigation. Gene Set Variation Analysis (GSVA) was performed to explore the biological pathways that were involved in subtypes. The chemo drug sensitivity and immunotherapy of subtypes were estimated through GDSC database and NTP algorithm. To detect EMT subtype-related prognostic gene modules, the analysis of weighted gene co-expression network (WGCNA) was performed. The LASSO algorithm was utilized to construct a prognostic risk model, and its efficacy was verified through an independent CGGA dataset. Finally, the expression of the hub genes from the prognostic model was evaluated through the single-cell dataset and in-vitro experiment. Results: The TCGA-LGG dataset revealed the creation of two molecular subtypes that presented different prognoses, clinical implications, TME, mutation landscapes, chemotherapy, and immunotherapy. A three-gene signature (SLC39A1, CTSA and CLIC1) based on EMT expression pattern were established through WGCNA analysis. Low-risk patients showed a positive outlook, increased immune cell presence, and higher expression of immune checkpoint proteins. In addition, several promising drugs, including birinapant, fluvastatin, clofarabine, dasatinib, tanespimycin, TAK-733, GDC-0152, AZD8330, trametinib and ingenol-mebutate had great potential to the treatment of high risk patients. Finally, CTSA and CLIC1 were highly expressed in monocyte cell through single-cell RNA sequencing analysis. Conclusion: Our research revealed non-negligible role of EMT in the TME diversity and complexity of LGG. A prognostic signature may contribute to the personalized treatment and prognostic determination.
Collapse
Affiliation(s)
- Chengcheng Wang
- Department of Pharmacy, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Zheng He
- Department of Neurosurgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| |
Collapse
|