1
|
Bhardwaj JS, Paliwal S, Singhvi G, Taliyan R. Immunological challenges and opportunities in glioblastoma multiforme: A comprehensive view from immune system lens. Life Sci 2024; 357:123089. [PMID: 39362586 DOI: 10.1016/j.lfs.2024.123089] [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: 01/28/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Glioblastoma multiforme (GBM), also known as grade IV astrocytoma, is the most common and deadly brain tumour. It has a poor prognosis and a low survival rate. GBM cells' immunological escape mechanism helps them resist advanced multimodal therapy. In physiological homeostasis, brain astrocytes and microglia suppress infections and clear the potential pathogen from the system. However, in severe pathological conditions like cancer, the immune response fails to eliminate mutated and rapidly over-proliferating GBM cells. The malignant cells' interactions with immune cells and the neoplasm's immunosuppressive environment enable the avoidance and their clearance. Immunotherapy efficiently addresses these difficulties, as shown by sufficient evidence. This review discusses how GBM cells inhibit and elude the immune system. These include MHC molecule expression alteration and PD-L1 and CTLA-4 immune checkpoint overexpression. Without co-stimulation, these changes induce effector T-cell tolerance and anergy. The review also covers how MDSCs, TAMs, Herpes Virus Entry Mediators, and Human cytomegalovirus protein decrease the effector immune response against glioblastoma. The latter part discusses various therapies that are available in the market or under clinical trials which revolves around combating resistance against the available multimodal therapies. The recent trends indicate that there are various monoclonal antibodies and peptide-based vaccines that can be utilized to overcome the immune evasion technique harbored by GBM cells. A strategic development of Immunotherapy considering these hallmarks of immune evasion may help in designing a therapy that may prove to be effective in killing the GBM cells thereby, improving the overall survival of GBM-affected patients.
Collapse
Affiliation(s)
- Jayant Singh Bhardwaj
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Shivangi Paliwal
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India.
| |
Collapse
|
2
|
Wang VG, Liu Z, Martinek J, Foroughi Pour A, Zhou J, Boruchov H, Ray K, Palucka K, Chuang JH. Computational immune synapse analysis reveals T-cell interactions in distinct tumor microenvironments. Commun Biol 2024; 7:1201. [PMID: 39341903 PMCID: PMC11438971 DOI: 10.1038/s42003-024-06902-2] [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: 03/11/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
The tumor microenvironment (TME) and the cellular interactions within it can be critical to tumor progression and treatment response. Although technologies to generate multiplex images of the TME are advancing, the many ways in which TME imaging data can be mined to elucidate cellular interactions are only beginning to be realized. Here, we present a novel approach for multipronged computational immune synapse analysis (CISA) that reveals T-cell synaptic interactions from multiplex images. CISA enables automated discovery and quantification of immune synapse interactions based on the localization of proteins on cell membranes. We first demonstrate the ability of CISA to detect T-cell:APC (antigen presenting cell) synaptic interactions in two independent human melanoma imaging mass cytometry (IMC) tissue microarray datasets. We then verify CISA's applicability across data modalities with melanoma histocytometry whole slide images, revealing that T-cell:macrophage synapse formation correlates with T-cell proliferation. We next show the generality of CISA by extending it to breast cancer IMC images, finding that CISA quantifications of T-cell:B-cell synapses are predictive of improved patient survival. Our work demonstrates the biological and clinical significance of spatially resolving cell-cell synaptic interactions in the TME and provides a robust method to do so across imaging modalities and cancer types.
Collapse
Affiliation(s)
- Victor G Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
| | - Zichao Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
| | - Jan Martinek
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Jie Zhou
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
| | - Hannah Boruchov
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Kelly Ray
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
| |
Collapse
|
3
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Pavlov KA, Chekhonin VP. Systemic and local immunosuppression in glioblastoma and its prognostic significance. Front Immunol 2024; 15:1326753. [PMID: 38481999 PMCID: PMC10932993 DOI: 10.3389/fimmu.2024.1326753] [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: 10/23/2023] [Accepted: 02/06/2024] [Indexed: 04/07/2024] Open
Abstract
The effectiveness of tumor therapy, especially immunotherapy and oncolytic virotherapy, critically depends on the activity of the host immune cells. However, various local and systemic mechanisms of immunosuppression operate in cancer patients. Tumor-associated immunosuppression involves deregulation of many components of immunity, including a decrease in the number of T lymphocytes (lymphopenia), an increase in the levels or ratios of circulating and tumor-infiltrating immunosuppressive subsets [e.g., macrophages, microglia, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs)], as well as defective functions of subsets of antigen-presenting, helper and effector immune cell due to altered expression of various soluble and membrane proteins (receptors, costimulatory molecules, and cytokines). In this review, we specifically focus on data from patients with glioblastoma/glioma before standard chemoradiotherapy. We discuss glioblastoma-related immunosuppression at baseline and the prognostic significance of different subsets of circulating and tumor-infiltrating immune cells (lymphocytes, CD4+ and CD8+ T cells, Tregs, natural killer (NK) cells, neutrophils, macrophages, MDSCs, and dendritic cells), including neutrophil-to-lymphocyte ratio (NLR), focus on the immune landscape and prognostic significance of isocitrate dehydrogenase (IDH)-mutant gliomas, proneural, classical and mesenchymal molecular subtypes, and highlight the features of immune surveillance in the brain. All attempts to identify a reliable prognostic immune marker in glioblastoma tissue have led to contradictory results, which can be explained, among other things, by the unprecedented level of spatial heterogeneity of the immune infiltrate and the significant phenotypic diversity and (dys)functional states of immune subpopulations. High NLR is one of the most repeatedly confirmed independent prognostic factors for shorter overall survival in patients with glioblastoma and carcinoma, and its combination with other markers of the immune response or systemic inflammation significantly improves the accuracy of prediction; however, more prospective studies are needed to confirm the prognostic/predictive power of NLR. We call for the inclusion of dynamic assessment of NLR and other blood inflammatory markers (e.g., absolute/total lymphocyte count, platelet-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, systemic immune-inflammation index, and systemic immune response index) in all neuro-oncology studies for rigorous evaluation and comparison of their individual and combinatorial prognostic/predictive significance and relative superiority.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Konstantin A. Pavlov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
4
|
Jaraíz-Rodríguez M, Del Prado L, Balsa E. Metabolic remodeling in astrocytes: Paving the path to brain tumor development. Neurobiol Dis 2023; 188:106327. [PMID: 37839712 DOI: 10.1016/j.nbd.2023.106327] [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: 06/28/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023] Open
Abstract
The brain is a highly metabolic organ, composed of multiple cell classes, that controls crucial functions of the body. Although neurons have traditionally been the main protagonist, astrocytes have gained significant attention over the last decade. In this regard, astrocytes are a type of glial cells that have recently emerged as critical regulators of central nervous system (CNS) function and play a significant role in maintaining brain energy metabolism. However, in certain scenarios, astrocyte behavior can go awry, which poses a significant threat to brain integrity and function. This is definitively the case for mutations that turn normal astrocytes and astrocytic precursors into gliomas, an aggressive type of brain tumor. In addition, healthy astrocytes can interact with tumor cells, becoming part of the tumor microenvironment and influencing disease progression. In this review, we discuss the recent evidence suggesting that disturbed metabolism in astrocytes can contribute to the development and progression of fatal human diseases such as cancer. Emphasis is placed on detailing the molecular bases and metabolic pathways of this disease and highlighting unique metabolic vulnerabilities that can potentially be exploited to develop successful therapeutic opportunities.
Collapse
Affiliation(s)
- Myriam Jaraíz-Rodríguez
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Lucia Del Prado
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Eduardo Balsa
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain; Instituto Universitario de Biología Molecular - IUBM (Universidad Autónoma de Madrid), Madrid, Spain.
| |
Collapse
|
5
|
Wang V, Liu Z, Martinek J, Zhou J, Boruchov H, Ray K, Palucka K, Chuang J. Computational immune synapse analysis reveals T-cell interactions in distinct tumor microenvironments. RESEARCH SQUARE 2023:rs.3.rs-2968528. [PMID: 37398220 PMCID: PMC10312981 DOI: 10.21203/rs.3.rs-2968528/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The tumor microenvironment (TME) and the cellular interactions within it can be critical to tumor progression and treatment response. Although technologies to generate multiplex images of the TME are advancing, the many ways in which TME imaging data can be mined to elucidate cellular interactions are only beginning to be realized. Here, we present a novel approach for multipronged computational immune synapse analysis (CISA) that reveals T-cell synaptic interactions from multiplex images. CISA enables automated discovery and quantification of immune synapse interactions based on the localization of proteins on cell membranes. We first demonstrate the ability of CISA to detect T-cell:APC (antigen presenting cell) synaptic interactions in two independent human melanoma imaging mass cytometry (IMC) tissue microarray datasets. We then generate melanoma histocytometry whole slide images and verify that CISA can detect similar interactions across data modalities. Interestingly, CISA histoctyometry analysis also reveals that T-cell:macrophage synapse formation is associated with T-cell proliferation. We next show the generality of CISA by extending it to breast cancer IMC images, finding that CISA quantifications of T-cell:B-cell synapses are predictive of improved patient survival. Our work demonstrates the biological and clinical significance of spatially resolving cell-cell synaptic interactions in the TME and provides a robust method to do so across imaging modalities and cancer types.
Collapse
Affiliation(s)
| | - Zichao Liu
- 1The Jackson Laboratory for Genomic Medicine
| | | | - Jie Zhou
- The Jackson Laboratory for Genomic Medicine
| | | | - Kelly Ray
- The Jackson Laboratory for Genomic Medicine
| | | | | |
Collapse
|
6
|
Rocha Pinheiro SL, Lemos FFB, Marques HS, Silva Luz M, de Oliveira Silva LG, Faria Souza Mendes dos Santos C, da Costa Evangelista K, Calmon MS, Sande Loureiro M, Freire de Melo F. Immunotherapy in glioblastoma treatment: Current state and future prospects. World J Clin Oncol 2023; 14:138-159. [PMID: 37124134 PMCID: PMC10134201 DOI: 10.5306/wjco.v14.i4.138] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023] Open
Abstract
Glioblastoma remains as the most common and aggressive malignant brain tumor, standing with a poor prognosis and treatment prospective. Despite the aggressive standard care, such as surgical resection and chemoradiation, median survival rates are low. In this regard, immunotherapeutic strategies aim to become more attractive for glioblastoma, considering its recent advances and approaches. In this review, we provide an overview of the current status and progress in immunotherapy for glioblastoma, going through the fundamental knowledge on immune targeting to promising strategies, such as Chimeric antigen receptor T-Cell therapy, immune checkpoint inhibitors, cytokine-based treatment, oncolytic virus and vaccine-based techniques. At last, it is discussed innovative methods to overcome diverse challenges, and future perspectives in this area.
Collapse
Affiliation(s)
- Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Hanna Santos Marques
- Campus Vitória da Conquista, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | | | - Mariana Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Matheus Sande Loureiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
7
|
Bispecific Antibody Format and the Organization of Immunological Synapses in T Cell-Redirecting Strategies for Cancer Immunotherapy. Pharmaceutics 2022; 15:pharmaceutics15010132. [PMID: 36678761 PMCID: PMC9863865 DOI: 10.3390/pharmaceutics15010132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023] Open
Abstract
T cell-redirecting strategies have emerged as effective cancer immunotherapy approaches. Bispecific antibodies (bsAbs) are designed to specifically recruit T cells to the tumor microenvironment and induce the assembly of the immunological synapse (IS) between T cells and cancer cells or antigen-presenting cells. The way that the quality of the IS might predict the effectiveness of T cell-redirecting strategies, including those mediated by bsAbs or by chimeric antigen receptors (CAR)-T cells, is currently under discussion. Here we review the organization of the canonical IS assembled during natural antigenic stimulation through the T cell receptor (TCR) and to what extent different bsAbs induce T cell activation, canonical IS organization, and effector function. Then, we discuss how the biochemical parameters of different formats of bsAbs affect the effectivity of generating an antigen-induced canonical IS. Finally, the quality of the IS assembled by bsAbs and monoclonal antibodies or CAR-T cells are compared, and strategies to improve bsAb-mediated T cell-redirecting strategies are discussed.
Collapse
|
8
|
Zhao Y, Zhu W, Chen H, Yan K, Wu J, Huang Q. Glioma stem cells and their microenvironment: A narrative review on docking and transformation. GLIOMA 2022. [DOI: 10.4103/glioma.glioma_5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
9
|
Haehnel S, Rade M, Kaiser N, Reiche K, Horn A, Loeffler D, Blumert C, Rapp F, Horn F, Meixensberger J, Renner C, Mueller W, Gaunitz F, Bechmann I, Winter K. RNA sequencing of glioblastoma tissue slice cultures reveals the effects of treatment at the transcriptional level. FEBS Open Bio 2021; 12:480-493. [PMID: 34923780 PMCID: PMC8804611 DOI: 10.1002/2211-5463.13353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 11/30/2022] Open
Abstract
One of the major challenges in cancer research is finding models that closely resemble tumors within patients. Human tissue slice cultures are a promising approach to provide a model of the patient's tumor biology ex vivo. Recently, it was shown that these slices can be successfully analyzed by whole transcriptome sequencing as well as automated histochemistry, increasing their usability as preclinical model. Glioblastoma multiforme (GBM) is a highly malignant brain tumor with poor prognosis and little is known about its genetic background and heterogeneity regarding therapy success. In this study, tissue from the tumors of 25 patients with primary GBM was processed into slice cultures and treated with standard therapy (irradiation and temozolomide). Total RNA sequencing and automated histochemistry were performed to enable analysis of treatment effects at a transcriptional and histological level. Slice cultures from long‐term survivors (overall survival [OS] > 24 months) exhibited more apoptosis than cultures from patients with shorter OS. Proliferation within these slices was slightly increased in contrast to other groups, but not significantly. Among all samples, 58 protein‐coding genes were upregulated and 32 downregulated in treated vs. untreated slice cultures. In general, an upregulation of DNA damage‐related and cell cycle checkpoint genes as well as enrichment of genotoxicity pathways and p53‐dependent signaling was found after treatment. Overall, the current study reproduces knowledge from former studies regarding the feasibility of transcriptomic analyses and automated histology in tissue slice cultures. We further demonstrate that the experimental data merge with the clinical follow‐up of the patients, which improves the applicability of our model system.
Collapse
Affiliation(s)
- Susann Haehnel
- Institute of Anatomy, Faculty of Medicine, University of Leipzig, Germany
| | - Michael Rade
- Department of Diagnostics, Fraunhofer Institute of Cell Therapy and Immunology, Leipzig, Germany
| | - Nicole Kaiser
- Institute of Anatomy, Faculty of Medicine, University of Leipzig, Germany
| | - Kristin Reiche
- Department of Diagnostics, Fraunhofer Institute of Cell Therapy and Immunology, Leipzig, Germany
| | - Andreas Horn
- Institute of Anatomy, Faculty of Medicine, University of Leipzig, Germany
| | - Dennis Loeffler
- Department of Diagnostics, Fraunhofer Institute of Cell Therapy and Immunology, Leipzig, Germany
| | - Conny Blumert
- Department of Diagnostics, Fraunhofer Institute of Cell Therapy and Immunology, Leipzig, Germany
| | - Felicitas Rapp
- GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Friedemann Horn
- Department of Diagnostics, Fraunhofer Institute of Cell Therapy and Immunology, Leipzig, Germany.,Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Germany
| | | | | | - Wolf Mueller
- Department of Neuropathology, University Hospital Leipzig, Germany
| | - Frank Gaunitz
- Department of Neurosurgery, University Hospital Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Faculty of Medicine, University of Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Faculty of Medicine, University of Leipzig, Germany
| |
Collapse
|
10
|
Hu J, Han C, Zhong J, Liu H, Liu R, Luo W, Chen P, Ling F. Dynamic Network Biomarker of Pre-Exhausted CD8 + T Cells Contributed to T Cell Exhaustion in Colorectal Cancer. Front Immunol 2021; 12:691142. [PMID: 34434188 PMCID: PMC8381053 DOI: 10.3389/fimmu.2021.691142] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has achieved positive clinical responses in various cancers. However, in advanced colorectal cancer (CRC), immunotherapy is challenging because of the deterioration of T-cell exhaustion, the mechanism of which is still unclear. In this study, we depicted CD8+ T-cell developmental trajectories and characterized the pre-exhausted T cells isolated from CRC patients in the scRNA-seq data set using a dynamic network biomarker (DNB). Moreover, CCT6A identified by DNB was a biomarker for pre-exhausted T-cell subpopulation in CRC. Besides, TUBA1B expression was triggered by CCT6A as DNB core genes contributing to CD8+ T cell exhaustion, indicating that core genes serve as biomarkers in pre-exhausted T cells. Remarkably, both TUBA1B and CCT6A expressions were significantly associated with the overall survival of COAD patients in the TCGA database (p = 0.0082 and p = 0.026, respectively). We also observed that cellular communication between terminally differentiated exhausted T cells and pre-exhausted T cells contributes to exhaustion. These findings provide new insights into the mechanism of T-cell exhaustion and provide clue for targeted immunotherapy in CRC.
Collapse
Affiliation(s)
- Jiaqi Hu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Chongyin Han
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jiayuan Zhong
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Huisheng Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Rui Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Wei Luo
- The First People's Hospital of Foshan, Sun Yat-sen University, Foshan, China
| | - Pei Chen
- School of Mathematics, South China University of Technology, Guangzhou, China.,Pazhou Lab, Guangzhou, China
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
11
|
Tietze S, Michen S, Schackert G, Temme A. Prospects of immune checkpoint blockade and vaccine-based immunotherapy for glioblastoma. Innov Surg Sci 2021. [DOI: 10.1515/iss-2020-0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
Glioblastoma multiforme (GBM) is the most prevalent primary brain tumor endowed with a dismal prognosis. Nowadays, immunotherapy in a particular immune checkpoint blockade and therapeutic vaccines are being extensively pursued. Yet, several characteristics of GBM may impact such immunotherapeutic approaches. This includes tumor heterogeneity, the relatively low mutational load of primary GBM, insufficient delivery of antibodies to tumor parenchyma and the unique immunosuppressive microenvironment of GBM. Moreover, standard treatment of GBM, comprising temozolomide chemotherapy, radiotherapy and in most instances the application of glucocorticoids for management of brain edema, results in a further increased immunosuppression. This review will provide a brief introduction to the principles of vaccine-based immunotherapy and give an overview of the current clinical studies, which employed immune checkpoint inhibitors, oncolytic viruses-based vaccination, cell-based and peptide-based vaccines. Recent experiences as well as the latest developments are reviewed. Overcoming obstacles, which limit the induction and long-term immune response against GBM when using vaccination approaches, are necessary for the implementation of effective immunotherapy of GBM.
Collapse
Affiliation(s)
- Stefanie Tietze
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology , University Hospital Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Susanne Michen
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology , University Hospital Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Gabriele Schackert
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology , University Hospital Carl Gustav Carus, Technical University Dresden , Dresden , Germany
- German Cancer Consortium (DKTK) , Dresden , Germany
- German Cancer Research Center (DKFZ) , Heidelberg , Germany
- National Center for Tumor Diseases , University Hospital Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Achim Temme
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology , University Hospital Carl Gustav Carus, Technical University Dresden , Dresden , Germany
- German Cancer Consortium (DKTK) , Dresden , Germany
- German Cancer Research Center (DKFZ) , Heidelberg , Germany
- National Center for Tumor Diseases , University Hospital Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| |
Collapse
|
12
|
Cribaro GP, Saavedra-López E, Romarate L, Mitxitorena I, Díaz LR, Casanova PV, Roig-Martínez M, Gallego JM, Perez-Vallés A, Barcia C. Three-dimensional vascular microenvironment landscape in human glioblastoma. Acta Neuropathol Commun 2021; 9:24. [PMID: 33579378 PMCID: PMC7879533 DOI: 10.1186/s40478-020-01115-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
The cellular complexity of glioblastoma microenvironments is still poorly understood. In-depth, cell-resolution tissue analyses of human material are rare but highly necessary to understand the biology of this deadly tumor. Here we present a unique 3D visualization revealing the cellular composition of human GBM in detail and considering its critical association with the neo-vascular niche. Our images show a complex vascular map of human 3D biopsies with increased vascular heterogeneity and altered spatial relationship with astrocytes or glioma-cell counterparts. High-resolution analysis of the structural layers of the blood brain barrier showed a multilayered fenestration of endothelium and basement membrane. Careful examination of T cell position and migration relative to vascular walls revealed increased infiltration corresponding with tumor proliferation. In addition, the analysis of the myeloid landscape not only showed a volumetric increase in glioma-associated microglia and macrophages relative to GBM proliferation but also revealed distinct phenotypes in tumor nest and stroma. Images and data sets are available on demand as a resource for public access.
Collapse
|
13
|
IL-21 Increases the Reactivity of Allogeneic Human Vγ9Vδ2 T Cells Against Primary Glioblastoma Tumors. J Immunother 2019; 41:224-231. [PMID: 29683891 DOI: 10.1097/cji.0000000000000225] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Glioblastoma multiforme (GBM) remains the most frequent and deadliest primary brain tumor in adults despite aggressive treatments, because of the persistence of infiltrative and resistant tumor cells. Nonalloreactive human Vγ9Vδ2 T lymphocytes, the major peripheral γδ T-cell subset in adults, represent attractive effectors for designing immunotherapeutic strategies to track and eliminate brain tumor cells, with limited side effects. We analyzed the effects of ex vivo sensitizations of Vγ9Vδ2 T cells by IL-21, a modulating cytokine, on their cytolytic reactivity. We first showed that primary human GBM-1 cells were naturally eliminated by allogeneic Vγ9Vδ2 T lymphocytes, through a perforin/granzyme-mediated cytotoxicity. IL-21 increased both intracellular granzyme B levels and cytotoxicity of allogeneic human Vγ9Vδ2 T lymphocytes in vitro. Importantly, IL-21-enhanced cytotoxicity was rapid, which supports the development of sensitization(s) of γδ T lymphocytes before adoptive transfer, a process that avoids any deleterious effect associated with direct administrations of IL-21. Finally, we showed, for the first time, that IL-21-sensitized allogeneic Vγ9Vδ2 T cells significantly eliminated GBM tumor cells that developed in the brain after orthotopic administrations in vivo. Altogether our observations pave the way for novel efficient stereotaxic immunotherapies in GBM patients by using IL-21-sensitized allogeneic human Vγ9Vδ2 T cells.
Collapse
|
14
|
Priego N, Valiente M. The Potential of Astrocytes as Immune Modulators in Brain Tumors. Front Immunol 2019; 10:1314. [PMID: 31244853 PMCID: PMC6579886 DOI: 10.3389/fimmu.2019.01314] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
The neuro-immune axis has emerged as a key aspect to understand the normal function of the Central Nervous System (CNS) as well as the pathophysiology of many brain disorders. As such, it may represent a promising source for novel therapeutic targets. Glial cells, and in particular the extensively studied microglia, play important roles in brain disorders. Astrocytes, in their reactive state, have been shown to positively and negatively modulate the progression of multiple CNS disorders. These seemingly opposing effects, might stem from their underlying heterogeneity, an aspect that has recently come to light. In this article we will discuss the link between reactive astrocytes and the neuro-immune axis with a perspective on their potential importance in brain tumors. Based on the gained knowledge from studies in other CNS disorders, reactive astrocytes are undoubtfully emerging as a key component of the neuro-immune axis, with ability to modulate both the innate and adaptive branches of the immune system. Lastly, we will discuss how we can exploit our improved understanding of the basic biology of astrocytes to further enhance the efficacy of emerging immune-based therapies in primary brain tumors and brain metastasis.
Collapse
Affiliation(s)
- Neibla Priego
- Brain Metastasis Group, Molecular Oncology Programme, National Cancer Research Center (CNIO), Madrid, Spain
| | - Manuel Valiente
- Brain Metastasis Group, Molecular Oncology Programme, National Cancer Research Center (CNIO), Madrid, Spain
| |
Collapse
|
15
|
Novel concept of the border niche: glioblastoma cells use oligodendrocytes progenitor cells (GAOs) and microglia to acquire stem cell-like features. Brain Tumor Pathol 2019; 36:63-73. [PMID: 30968276 DOI: 10.1007/s10014-019-00341-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 03/23/2019] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is a major malignant brain tumor developing in adult brain white matter, characterized by rapid growth and invasion. GBM cells spread into the contralateral hemisphere, even during early tumor development. However, after complete resection of tumor mass, GBM commonly recurs around the tumor removal cavity, suggesting that a microenvironment at the tumor border provides chemo-radioresistance to GBM cells. Thus, clarification of the tumor border microenvironment is critical for improving prognosis in GBM patients. MicroRNA (miRNA) expression in samples from the tumor, tumor border, and peripheral region far from tumor mass was compared, and five miRNAs showing characteristically higher expression in the tumor border were identified, with the top three related to oligodendrocyte differentiation. Pathologically, oligodendrocyte lineage cells increased in the border, but were rare in tumors. Macrophages/microglia also colocalized in the border area. Medium cultured with oligodendrocyte progenitor cells (OPCs) and macrophages induced stemness and chemo-radioresistance in GBM cells, suggesting that OPCs and macrophages/microglia constitute a special microenvironment for GBM cells at the tumor border. The supportive function of OPCs for GBM cells has not been discussed previously. OPCs are indispensable for GBM cells to establish special niches for chemo-radioresistance outside the tumor mass.
Collapse
|
16
|
Congdon KL, Sanchez-Perez LA, Sampson JH. Effective effectors: How T cells access and infiltrate the central nervous system. Pharmacol Ther 2018; 197:52-60. [PMID: 30557632 DOI: 10.1016/j.pharmthera.2018.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Several Phase II and III clinical trials have demonstrated that immunotherapy can induce objective responses in otherwise refractory malignancies in tumors outside the central nervous system. In large part, effector T cells mediate much of the antitumor efficacy in these trials, and potent antitumor T cells can be generated through vaccination, immune checkpoint blockade, adoptive transfer, and genetic manipulation. However, activated T cells must still traffic to, infiltrate, and persist within tumor in order to mediate tumor lysis. These requirements for efficacy pose unique challenges for brain tumor immunotherapy, due to specific anatomical barriers and populations of specialized immune cells within the central nervous system that function to constrain immunity. Both autoimmune and infectious diseases of the central nervous system provide a wealth of information on how T cells can successfully migrate to the central nervous system and then engender sustained immune responses. In this review, we will examine the commonalities in the efferent arm of immunity to the brain for autoimmunity, infection, and tumor immunotherapy to identify key factors underlying potent immune responses.
Collapse
Affiliation(s)
- Kendra L Congdon
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, United States; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, United States
| | - Luis A Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, United States; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, United States
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, United States; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, United States; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, United States; Department of Pathology, Duke University School of Medicine, Durham, NC 27710, United States.
| |
Collapse
|
17
|
Díaz LR, Saavedra-López E, Romarate L, Mitxitorena I, Casanova PV, Cribaro GP, Gallego JM, Pérez-Vallés A, Forteza-Vila J, Alfaro-Cervello C, García-Verdugo JM, Barcia C, Barcia C. Imbalance of immunological synapse-kinapse states reflects tumor escape to immunity in glioblastoma. JCI Insight 2018; 3:120757. [PMID: 30232280 DOI: 10.1172/jci.insight.120757] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/17/2018] [Indexed: 12/19/2022] Open
Abstract
Since the proper activation of T cells requires the physical interaction with target cells through the formation of immunological synapses (IS), an alteration at this level could be a reason why tumors escape the immune response. As part of their life cycle, it is thought that T cells alternate between a static phase, the IS, and a dynamic phase, the immunological kinapse (IK), depending on high or low antigen sensing. Our investigation performed in tissue samples of human glioma shows that T cells are able to establish synapsing interactions not only with glioma tumorigenic cells, but also with stromal myeloid cells. Particularly, the IS displaying a T cell receptor-rich (TCR-rich) central supramolecular activation cluster (cSMAC) is preferentially established with stromal cells, as opposed to malignant cells. Conversely, T cells in the malignant areas showed distinct morphometric parameters compared with nonneoplastic tissue - the former characterized by an elongated shape, well-suited to kinaptic dynamics. Importantly, high-resolution 3-dimensional analyses demonstrated the existence of bona-fide IK preferentially arranged in malignant areas of the tumor. This imbalance of IS/IK states between these 2 microenvironments reveals the low antigenic sensing of T cells when patrolling tumorigenic cells and reflects the immunoevasive environment of the tumor.
Collapse
Affiliation(s)
- Laura R Díaz
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Elena Saavedra-López
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Leire Romarate
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Izaskun Mitxitorena
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - Paola V Casanova
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | - George P Cribaro
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| | | | - Ana Pérez-Vallés
- Department of Pathology, Valencia General Hospital, Valencia, Spain
| | - Jerónimo Forteza-Vila
- Unidad Mixta CIPF/UCV de Investigación Oncológica, Instituto Valenciano de Patología, Universidad Católica de Valencia, Valencia, Spain
| | - Clara Alfaro-Cervello
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universitat de València, CIBERNED, Valencia, Spain
| | - José M García-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universitat de València, CIBERNED, Valencia, Spain
| | | | - Carlos Barcia
- Department of Biochemistry and Molecular Biology, School of Medicine, and.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
18
|
Martinvalet D. The role of the mitochondria and the endoplasmic reticulum contact sites in the development of the immune responses. Cell Death Dis 2018; 9:336. [PMID: 29491398 PMCID: PMC5832423 DOI: 10.1038/s41419-017-0237-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/25/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022]
Abstract
Mitochondria and endoplasmic reticulum (ER) contact sites (MERCs) are dynamic modules enriched in subset of lipids and specialized proteins that determine their structure and functions. The MERCs regulate lipid transfer, autophagosome formation, mitochondrial fission, Ca2+ homeostasis and apoptosis. Since these functions are essential for cell biology, it is therefore not surprising that MERCs also play a critical role in organ physiology among which the immune system stands by its critical host defense function. This defense system must discriminate and tolerate host cells and beneficial commensal microorganisms while eliminating pathogenic ones in order to preserve normal homeostasis. To meet this goal, the immune system has two lines of defense. First, the fast acting but unspecific innate immune system relies on anatomical physical barriers and subsets of hematopoietically derived cells expressing germline-encoded receptors called pattern recognition receptors (PRR) recognizing conserved motifs on the pathogens. Second, the slower but very specific adaptive immune response is added to complement innate immunity. Adaptive immunity relies on another set of specialized cells, the lymphocytes, harboring receptors requiring somatic recombination to be expressed. Both innate and adaptive immune cells must be activated to phagocytose and process pathogens, migrate, proliferate, release soluble factors and destroy infected cells. Some of these functions are strongly dependent on lipid transfer, autophagosome formation, mitochondrial fission, and Ca2+ flux; this indicates that MERCs could regulate immunity.
Collapse
Affiliation(s)
- Denis Martinvalet
- Department of Cell Physiology and Metabolism, Geneva Medical School, 1211, Geneva, Switzerland.
| |
Collapse
|
19
|
Johanns TM, Bowman-Kirigin JA, Liu C, Dunn GP. Targeting Neoantigens in Glioblastoma: An Overview of Cancer Immunogenomics and Translational Implications. Neurosurgery 2017; 64:165-176. [PMID: 28899059 PMCID: PMC6287409 DOI: 10.1093/neuros/nyx321] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/27/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
- Tanner M. Johanns
- Division of Oncology, Department of Medicine, Washington University School of
Medicine, St. Louis, Missouri
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington
Univer-sity School of Medicine, St. Louis, Missouri
| | - Jay A. Bowman-Kirigin
- Center for Human Immunology and Immunotherapy Prog-rams, Washington University
School of Medicine, St. Louis, Missouri
- Depart-ment of Neurological Surgery, Washing-ton University School of Medicine,
St. Louis, Missouri
| | - Connor Liu
- Center for Human Immunology and Immunotherapy Prog-rams, Washington University
School of Medicine, St. Louis, Missouri
- Depart-ment of Neurological Surgery, Washing-ton University School of Medicine,
St. Louis, Missouri
| | - Gavin P. Dunn
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington
Univer-sity School of Medicine, St. Louis, Missouri
- Depart-ment of Neurological Surgery, Washing-ton University School of Medicine,
St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of
Medicine, St. Louis, Missouri
| |
Collapse
|
20
|
Bassoy EY, Kasahara A, Chiusolo V, Jacquemin G, Boydell E, Zamorano S, Riccadonna C, Pellegatta S, Hulo N, Dutoit V, Derouazi M, Dietrich PY, Walker PR, Martinvalet D. ER-mitochondria contacts control surface glycan expression and sensitivity to killer lymphocytes in glioma stem-like cells. EMBO J 2017; 36:1493-1512. [PMID: 28283580 DOI: 10.15252/embj.201695429] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma is a highly heterogeneous aggressive primary brain tumor, with the glioma stem-like cells (GSC) being more sensitive to cytotoxic lymphocyte-mediated killing than glioma differentiated cells (GDC). However, the mechanism behind this higher sensitivity is unclear. Here, we found that the mitochondrial morphology of GSCs modulates the ER-mitochondria contacts that regulate the surface expression of sialylated glycans and their recognition by cytotoxic T lymphocytes and natural killer cells. GSCs displayed diminished ER-mitochondria contacts compared to GDCs. Forced ER-mitochondria contacts in GSCs increased their cell surface expression of sialylated glycans and reduced their susceptibility to cytotoxic lymphocytes. Therefore, mitochondrial morphology and dynamism dictate the ER-mitochondria contacts in order to regulate the surface expression of certain glycans and thus play a role in GSC recognition and elimination by immune effector cells. Targeting the mitochondrial morphology, dynamism, and contacts with the ER could be an innovative strategy to deplete the cancer stem cell compartment to successfully treat glioblastoma.
Collapse
Affiliation(s)
- Esen Yonca Bassoy
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Atsuko Kasahara
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.,Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Valentina Chiusolo
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Guillaume Jacquemin
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Emma Boydell
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Sebastian Zamorano
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Cristina Riccadonna
- Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Serena Pellegatta
- Department of Molecular Neuro-Oncology, Fondazione I.R.C.C.S. Istituto Neurologico C. Besta, Milan, Italy
| | - Nicolas Hulo
- Biomathematical and Biostatistical Analysis, Institute of Genetics and Genomics University of Geneva, Geneva, Switzerland
| | - Valérie Dutoit
- Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Madiha Derouazi
- Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Amal Therapeutics, Geneva, Switzerland
| | - Pierre Yves Dietrich
- Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Paul R Walker
- Department of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Denis Martinvalet
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
21
|
Abstract
In this chapter, we describe the technical details to visualize and analyze effector immunological synapses between T cells and astrocytes in the brain with high-resolution confocal imaging. This procedure is critical for the optimal and even penetration of labeling antibodies within the nerve tissue to obtain accurate staining and allow a uniform three-dimensional analysis of the T cell-astrocyte interactions. We emphasize here the comprehensive exploration of the tissue and analysis with confocal microscope as well as the display of microanatomical details of the three-dimensional reconstruction for interface visualization (including peripheral and central supramolecular activation clusters, effector molecules, and other organelles such as microtubule organizing centers (MTOCs) and Golgi apparatus).
Collapse
|
22
|
Prognostic role of tumour-infiltrating inflammatory cells in brain tumours: literature review. Curr Opin Neurol 2016; 28:647-58. [PMID: 26402405 DOI: 10.1097/wco.0000000000000251] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Both primary and metastatic brain tumours pose a significant and unmet clinical need. Immune cells infiltrating the tumour have been shown to affect the clinical course of various extracranial tumour types, but there is little knowledge on the role of tumour-infiltrating immune cells in brain tumours. Thus, the aim of this review was to recapitulate the reports on immune infiltrates in brain tumours and their prognostic significance. RECENT FINDINGS Immune infiltrates composed of various lymphocyte subsets and microglia/macrophages are frequently observed in brain tumours; however, their density and prognostic role seem to differ between tumour types. Central nervous system (CNS) metastases, particularly of melanoma, lung cancer and renal cell cancer, commonly show high amounts of tumour-infiltrating lymphocytes and tumour-infiltrating lymphocytes density strongly correlate with patient's overall survival times in patients with CNS metastases. In gliomas and primary CNS lymphomas, some studies also suggest a prognostic role of immune cell infiltration; however, methodological issues such as low sample size and retrospective study designs with heterogeneous patient populations preclude definite conclusions. Meningiomas typically harbour inflammatory infiltrates, but their correlation with the clinical course is unclear because of the lack of studies correlating immune cell infiltrates with outcome parameters. SUMMARY The available literature suggests a relevant role of immune infiltrates in the clinical course of some brain tumour types; however, further studies are required to better understand the interaction of the immune system and CNS neoplasms and to explore therapeutic opportunities with immunotherapies such as vaccines or immune checkpoint modulators.
Collapse
|
23
|
Glioma Stemlike Cells Enhance the Killing of Glioma Differentiated Cells by Cytotoxic Lymphocytes. PLoS One 2016; 11:e0153433. [PMID: 27073883 PMCID: PMC4830556 DOI: 10.1371/journal.pone.0153433] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/29/2016] [Indexed: 01/16/2023] Open
Abstract
Glioblastoma multiforme, the most aggressive primary brain tumor, is maintained by a subpopulation of glioma cells with self-renewal properties that are able to recapitulate the entire tumor even after surgical resection or chemo-radiotherapy. This typifies the vast heterogeneity of this tumor with the two extremes represented on one end by the glioma stemlike cells (GSC) and on the other by the glioma differentiated cells (GDC). Interestingly, GSC are more sensitive to immune effector cells than the GDC counterpart. However, how GSC impact on the killing on the GDC and vice versa is not clear. Using a newly developed cytotoxicity assay allowing to simultaneously monitor cytotoxic lymphocytes-mediated killing of GSC and GDC, we found that although GSC were always better killed and that their presence enhanced the killing of GDC. In contrast, an excess of GDC had a mild protective effect on the killing of GSC, depending on the CTL type. Overall, our results suggest that during combination therapy, immunotherapy would be the most effective after prior treatment with conventional therapies.
Collapse
|
24
|
Sánchez-Osuna M, Martínez-Escardó L, Granados-Colomina C, Martínez-Soler F, Pascual-Guiral S, Iglesias-Guimarais V, Velasco R, Plans G, Vidal N, Tortosa A, Barcia C, Bruna J, Yuste VJ. An intrinsic DFF40/CAD endonuclease deficiency impairs oligonucleosomal DNA hydrolysis during caspase-dependent cell death: a common trait in human glioblastoma cells. Neuro Oncol 2016; 18:950-61. [PMID: 26755073 DOI: 10.1093/neuonc/nov315] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/02/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) or grade IV astrocytoma is one of the most devastating human cancers. The loss of DFF40/CAD, the key endonuclease that triggers oligonucleosomal DNA fragmentation during apoptosis, has been linked to genomic instability and cell survival after radiation. Despite the near inevitability of GBM tumor recurrence after treatment, the relationship between DFF40/CAD and GBM remains unexplored. METHODS We studied the apoptotic behavior of human GBM-derived cells after apoptotic insult. We analyzed caspase activation and the protein levels and subcellular localization of DFF40/CAD apoptotic endonuclease. DFF40/CAD was also evaluated in histological sections from astrocytic tumors and nontumoral human brain. RESULTS We showed that GBM cells undergo incomplete apoptosis without generating oligonucleosomal DNA degradation despite the correct activation of executioner caspases. The major defect of GBM cells relied on the improper accumulation of DFF40/CAD at the nucleoplasmic subcellular compartment. Supporting this finding, DFF40/CAD overexpression allowed GBM cells to display oligonucleosomal DNA degradation after apoptotic challenge. Moreover, the analysis of histological slices from astrocytic tumors showed that DFF40/CAD immunoreactivity in tumoral GFAP-positive cells was markedly reduced when compared with nontumoral samples. CONCLUSIONS Our data highlight the low expression levels of DFF40/CAD and the absence of DNA laddering as common molecular traits in GBM. These findings could be of major importance for understanding the malignant behavior of remaining tumor cells after radiochemotherapy.
Collapse
Affiliation(s)
- María Sánchez-Osuna
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Laura Martínez-Escardó
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Carla Granados-Colomina
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Fina Martínez-Soler
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Sònia Pascual-Guiral
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Victoria Iglesias-Guimarais
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Roser Velasco
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Gerard Plans
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Noemi Vidal
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Avelina Tortosa
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Carlos Barcia
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Jordi Bruna
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| | - Victor J Yuste
- Cell Death, Senescence and Survival group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (M.S.-O., L.M.-E., C.G.-C., S.P.-G., V.I.-G., V.J.Y.); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (C.I.B.E.R.N.E.D.), Barcelona, Spain (M.S.-O., V.I.-G., V.J.Y.); Department of Basic Nursing, Institut d'Investigació Biomèdica de Bellvitge-Universitat de Barcelona, Barcelona, Spain (F.M.-S., A.T.); Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-I.C.O Duran i Reynals, Barcelona, Spain (R.V., G.P., N.V., J.B.); Group of Neuroplasticity and Regeneration (C.I.B.E.R.N.E.D.), Department of Cell Biology, Physiology and Immunology & Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain (R.V., J.B.); Neuropathology Institute, Hospital Universitari de Bellvitge, Barcelona, Spain (N.V.); Neuro-Immunity Group, Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (C.B.)
| |
Collapse
|
25
|
Renner DN, Malo CS, Jin F, Parney IF, Pavelko KD, Johnson AJ. Improved Treatment Efficacy of Antiangiogenic Therapy when Combined with Picornavirus Vaccination in the GL261 Glioma Model. Neurotherapeutics 2016; 13:226-36. [PMID: 26620211 PMCID: PMC4720676 DOI: 10.1007/s13311-015-0407-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The addition of antiangiogenic therapy to the standard-of-care treatment regimen for recurring glioblastoma has provided some clinical benefits while also delineating numerous caveats, prompting evaluation of the elicited alterations to the tumor microenvironment. Of critical importance, given the steadily increasing incorporation of immunotherapeutic approaches clinically, is an enhanced understanding of the interplay between angiogenic and immune response pathways within tumors. In the present study, the GL261 glioma mouse model was used to determine the effects of antiangiogenic treatment in an immune-competent host. Following weekly systemic administration of aflibercept, an inhibitor of vascular endothelial growth factor, tumor volume was assessed by magnetic resonance imaging and changes to the tumor microenvironment were determined. Treatment with aflibercept resulted in reduced tumor burden and increased survival compared with controls. Additionally, decreased vascular permeability and preservation of the integrity of tight junction proteins were observed. Treated tumors also displayed hallmarks of anti-angiogenic evasion, including marked upregulation of vascular endothelial growth factor expression and increased tumor invasiveness. Aflibercept was then administered in combination with a picornavirus-based antitumor vaccine and tumor progression was evaluated. This combination therapy significantly delayed tumor progression and extended survival beyond that observed for either therapy alone. As such, this work demonstrates the efficacy of combined antiangiogenic and immunotherapy approaches for treating established gliomas and provides a foundation for further evaluation of the effects of antiangiogenic therapy in the context of endogenous or vaccine-induced inflammatory responses.
Collapse
Affiliation(s)
- Danielle N Renner
- Neurobiology of Disease Graduate Program, Mayo Clinic, Rochester, MN, USA
| | | | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Ian F Parney
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
| | | | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
26
|
Schijns VEJC, Pretto C, Devillers L, Pierre D, Hofman FM, Chen TC, Mespouille P, Hantos P, Glorieux P, Bota DA, Stathopoulos A. First clinical results of a personalized immunotherapeutic vaccine against recurrent, incompletely resected, treatment-resistant glioblastoma multiforme (GBM) tumors, based on combined allo- and auto-immune tumor reactivity. Vaccine 2015; 33:2690-6. [PMID: 25865468 PMCID: PMC10494870 DOI: 10.1016/j.vaccine.2015.03.095] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/10/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) patients have a poor prognosis. After tumor recurrence statistics suggest an imminent death within 1-4.5 months. Supportive preclinical data, from a rat model, provided the rational for a prototype clinical vaccine preparation, named Gliovac (or ERC 1671) composed of autologous antigens, derived from the patient's surgically removed tumor tissue, which is administered together with allogeneic antigens from glioma tissue resected from other GBM patients. We now report the first results of the Gliovac treatment for treatment-resistant GBM patients. Nine (9) recurrent GBM patients, after standard of care treatment, including surgery radio- and chemotherapy temozolomide, and for US patients, also bevacizumab (Avastin™), were treated under a compassionate use/hospital exemption protocol. Gliovac was given intradermally, together with human GM-CSF (Leukine(®)), and preceded by a regimen of regulatory T cell-depleting, low-dose cyclophosphamide. Gliovac administration in patients that have failed standard of care therapies showed minimal toxicity and enhanced overall survival (OS). Six-month (26 weeks) survival for the nine Gliovac patients was 100% versus 33% in control group. At week 40, the published overall survival was 10% if recurrent, reoperated patients were not treated. In the Gliovac treated group, the survival at 40 weeks was 77%. Our data suggest that Gliovac has low toxicity and a promising efficacy. A phase II trial has recently been initiated in recurrent, bevacizumab naïve GBM patients (NCT01903330).
Collapse
Affiliation(s)
- Virgil E J C Schijns
- Cell Biology & Immunology Group, Wageningen University, PO Box 338, 6700 AH Wageningen, The Netherlands; Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands.
| | - Chrystel Pretto
- Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands
| | - Laurent Devillers
- Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands
| | - Denis Pierre
- Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands
| | - Florence M Hofman
- Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | - Thomas C Chen
- Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA; Epitopoietic Research Corporation (ERC), 1055 E Colorado Blvd., Suite 500, Pasadena, CA 91106, USA
| | | | - Peter Hantos
- Department of Neurosurgery, Arlon and Libramont Hospital, Arlon and Libramont, Belgium
| | | | - Daniela A Bota
- Department of Neurology/Neurosurgery, University of California at Irvine, UC Irvine Medical Center, Irvine, CA, USA
| | - Apostolos Stathopoulos
- Cell Biology & Immunology Group, Wageningen University, PO Box 338, 6700 AH Wageningen, The Netherlands; Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands; Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA; Epitopoietic Research Corporation (ERC), 1055 E Colorado Blvd., Suite 500, Pasadena, CA 91106, USA; Department of Neurosurgery, Arlon and Libramont Hospital, Arlon and Libramont, Belgium; Department of Neurology/Neurosurgery, University of California at Irvine, UC Irvine Medical Center, Irvine, CA, USA.
| |
Collapse
|
27
|
Renner DN, Jin F, Litterman AJ, Balgeman AJ, Hanson LM, Gamez JD, Chae M, Carlson BL, Sarkaria JN, Parney IF, Ohlfest JR, Pirko I, Pavelko KD, Johnson AJ. Effective Treatment of Established GL261 Murine Gliomas through Picornavirus Vaccination-Enhanced Tumor Antigen-Specific CD8+ T Cell Responses. PLoS One 2015; 10:e0125565. [PMID: 25933216 PMCID: PMC4416934 DOI: 10.1371/journal.pone.0125565] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/25/2015] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma (GBM) is among the most invasive and lethal of cancers, frequently infiltrating surrounding healthy tissue and giving rise to rapid recurrence. It is therefore critical to establish experimental model systems and develop therapeutic approaches that enhance anti-tumor immunity. In the current study, we have employed a newly developed murine glioma model to assess the efficacy of a novel picornavirus vaccination approach for the treatment of established tumors. The GL261-Quad system is a variation of the GL261 syngeneic glioma that has been engineered to expresses model T cell epitopes including OVA257-264. MRI revealed that both GL261 and GL261-Quad tumors display characteristic features of human gliomas such as heterogeneous gadolinium leakage and larger T2 weighted volumes. Analysis of brain-infiltrating immune cells demonstrated that GL261-Quad gliomas generate detectable CD8+ T cell responses toward the tumor-specific Kb:OVA257-264 antigen. Enhancing this response via a single intracranial or peripheral vaccination with picornavirus expressing the OVA257-264 antigen increased anti-tumor CD8+ T cells infiltrating the brain, attenuated progression of established tumors, and extended survival of treated mice. Importantly, the efficacy of the picornavirus vaccination is dependent on functional cytotoxic activity of CD8+ T cells, as the beneficial response was completely abrogated in mice lacking perforin expression. Therefore, we have developed a novel system for evaluating mechanisms of anti-tumor immunity in vivo, incorporating the GL261-Quad model, 3D volumetric MRI, and picornavirus vaccination to enhance tumor-specific cytotoxic CD8+ T cell responses and track their effectiveness at eradicating established gliomas in vivo.
Collapse
Affiliation(s)
- Danielle N. Renner
- Neurobiology of Disease Graduate Program, Mayo Clinic, Rochester, MN, United States of America
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Adam J. Litterman
- Department of Neurosurgery, University of Minnesota, Minneapolis MN, United States of America
| | - Alexis J. Balgeman
- Summer Undergraduate Research Fellowship, Mayo Clinic, Rochester, MN, United States of America
| | - Lisa M. Hanson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Jeffrey D. Gamez
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
| | - Michael Chae
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States of America
| | - Brett L. Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Ian F. Parney
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States of America
| | - John R. Ohlfest
- Department of Neurosurgery, University of Minnesota, Minneapolis MN, United States of America
| | - Istvan Pirko
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
| | - Kevin D. Pavelko
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail: (AJJ); (KDP)
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail: (AJJ); (KDP)
| |
Collapse
|
28
|
Han J, Alvarez-Breckenridge CA, Wang QE, Yu J. TGF-β signaling and its targeting for glioma treatment. Am J Cancer Res 2015; 5:945-955. [PMID: 26045979 PMCID: PMC4449428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 02/19/2015] [Indexed: 06/04/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a pleiotropic cytokine, secreted by a variety of cells including immune cells, tumor cells, and stromal cells. TGF-β signaling is dysregulated in cancer patients, and this aberrant signaling at least in part contributes to initiation and progression of many cancers including glioma. The dysregulated signaling components provide molecular targets for the treatment of glioma. In this article, we review TGF-β signaling and its targeting in glioma.
Collapse
Affiliation(s)
- Jianfeng Han
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State UniversityColumbus, Ohio 43210, USA
- The Ohio State University Comprehensive Cancer CenterColumbus, Ohio 43210, USA
| | | | - Qi-En Wang
- The Ohio State University Comprehensive Cancer CenterColumbus, Ohio 43210, USA
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State UniversityColumbus, Ohio 43210, USA
- The Ohio State University Comprehensive Cancer CenterColumbus, Ohio 43210, USA
| |
Collapse
|
29
|
Kupfer-type immunological synapses in vivo: Raison D'être of SMAC. Immunol Cell Biol 2014; 93:51-6. [PMID: 25267483 DOI: 10.1038/icb.2014.80] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/01/2014] [Accepted: 08/03/2014] [Indexed: 01/04/2023]
Abstract
T cells engage with antigen-presenting cells to form immunological synapses. These intimate contacts are characterized by the complex arrangement of molecules at the intercellular interface, which has been described as the supramolecular activation cluster (SMAC). However, due to T cells functioning without SMAC formation and the difficulties of studying these complex arrangements in vivo, its biological importance has been questioned. In light of recent data, we focus this review on the putative functionality of SMACs in T-cell synaptic contacts in vivo and emphasize the therapeutic potential of SMAC manipulation in immune-driven diseases.
Collapse
|
30
|
Olin MR, Pluhar GE, Andersen BM, Shaver R, Waldron NN, Moertel CL. Victory and defeat in the induction of a therapeutic response through vaccine therapy for human and canine brain tumors: a review of the state of the art. Crit Rev Immunol 2014; 34:399-432. [PMID: 25404047 PMCID: PMC4485925 DOI: 10.1615/critrevimmunol.2014011577] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Anti-tumor immunotherapy using tumor lysate-based vaccines has made great advances over recent decades. Cancer vaccines aim to elicit adaptive immune responses through various pathways by providing tumor and tumor-associated antigens with an immune stimulant or adjuvant. These anti-tumor vaccines are therefore developed as personalized treatments. Utilizing tumors as a source of vaccine antigens in immunotherapy has demonstrated promising results with minimal toxicity. However, to date, researchers have failed to overcome the overpowering immune suppressive effects within the tumor microenvironment. Immune suppression occurs naturally via multiple mechanisms. These mechanisms serve an important homeostatic role restoring a normal tissue microenvironment following an inflammatory response. Due to these suppressive mechanisms and the inherent heterogeneity of tumors, it is imperative to then elicit and maintain a specific tumoricidal response if vaccine therapy or some other combination of reagents is chosen. In this review, we focus on the historical use of tumors as a source of antigens to elicit a tumoricidal response and the limitations encountered that prevent greater success in immunotherapy. We describe the advantages and disadvantages of various vaccines and their ineffectiveness due to tumor-induced immune suppression.
Collapse
Affiliation(s)
- Michael R. Olin
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - G. Elizabeth Pluhar
- Department of Veterinary Medicine, College of Veterinary Medicine. University of Minnesota, St. Paul, MN 55108
| | - Brian M. Andersen
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - Rob Shaver
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - Nate N. Waldron
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | | |
Collapse
|
31
|
Barcia C, Mitxitorena I, Carrillo-de Sauvage MA, Gallego JM, Pérez-Vallés A, Barcia C. Imaging the microanatomy of astrocyte-T-cell interactions in immune-mediated inflammation. Front Cell Neurosci 2013; 7:58. [PMID: 23641198 PMCID: PMC3639405 DOI: 10.3389/fncel.2013.00058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/15/2013] [Indexed: 11/19/2022] Open
Abstract
The role of astrocytes in the immune-mediated inflammatory response in the brain is more prominent than previously thought. Astrocytes become reactive in response to neuro-inflammatory stimuli through multiple pathways, contributing significantly to the machinery that modifies the parenchymal environment. In particular, astrocytic signaling induces the establishment of critical relationships with infiltrating blood cells, such as lymphocytes, which is a fundamental process for an effective immune response. The interaction between astrocytes and T-cells involves complex modifications to both cell types, which undergo micro-anatomical changes and the redistribution of their binding and secretory domains. These modifications are critical for different immunological responses, such as for the effectiveness of the T-cell response, for the specific infiltration of these cells and their homing in the brain parenchyma, and for their correct apposition with antigen-presenting cells (APCs) to form immunological synapses (ISs). In this article, we review the current knowledge of the interactions between T-cells and astrocytes in the context of immune-mediated inflammation in the brain, based on the micro-anatomical imaging of these appositions by high-resolution confocal microscopy and three-dimensional rendering. The study of these dynamic interactions using detailed technical approaches contributes to understanding the function of astrocytes in inflammatory responses and paves the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Carlos Barcia
- Department of Neurosurgery, Hospital General Universitari de València València, Spain
| | | | | | | | | | | |
Collapse
|
32
|
O'Brien ER, Howarth C, Sibson NR. The role of astrocytes in CNS tumors: pre-clinical models and novel imaging approaches. Front Cell Neurosci 2013; 7:40. [PMID: 23596394 PMCID: PMC3627137 DOI: 10.3389/fncel.2013.00040] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 03/26/2013] [Indexed: 02/06/2023] Open
Abstract
Brain metastasis is a significant clinical problem, yet the mechanisms governing tumor cell extravasation across the blood-brain barrier (BBB) and CNS colonization are unclear. Astrocytes are increasingly implicated in the pathogenesis of brain metastasis but in vitro work suggests both tumoricidal and tumor-promoting roles for astrocyte-derived molecules. Also, the involvement of astrogliosis in primary brain tumor progression is under much investigation. However, translation of in vitro findings into in vivo and clinical settings has not been realized. Increasingly sophisticated resources, such as transgenic models and imaging technologies aimed at astrocyte-specific markers, will enable better characterization of astrocyte function in CNS tumors. Techniques such as bioluminescence and in vivo fluorescent cell labeling have potential for understanding the real-time responses of astrocytes to tumor burden. Transgenic models targeting signaling pathways involved in the astrocytic response also hold great promise, allowing translation of in vitro mechanistic findings into pre-clinical models. The challenging nature of in vivo CNS work has slowed progress in this area. Nonetheless, there has been a surge of interest in generating pre-clinical models, yielding insights into cell extravasation across the BBB, as well as immune cell recruitment to the parenchyma. While the function of astrocytes in the tumor microenvironment is still unknown, the relationship between astrogliosis and tumor growth is evident. Here, we review the role of astrogliosis in both primary and secondary brain tumors and outline the potential for the use of novel imaging modalities in research and clinical settings. These imaging approaches have the potential to enhance our understanding of the local host response to tumor progression in the brain, as well as providing new, more sensitive diagnostic imaging methods.
Collapse
Affiliation(s)
- Emma R. O'Brien
- Department of Oncology, CR-UK/MRC Gray Institute for Radiation Oncology and Biology, Churchill Hospital, University of OxfordOxford, UK
| | | | | |
Collapse
|
33
|
Ohlfest JR, Andersen BM, Litterman AJ, Xia J, Pennell CA, Swier LE, Salazar AM, Olin MR. Vaccine injection site matters: qualitative and quantitative defects in CD8 T cells primed as a function of proximity to the tumor in a murine glioma model. THE JOURNAL OF IMMUNOLOGY 2012; 190:613-20. [PMID: 23248259 DOI: 10.4049/jimmunol.1201557] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Malignant gliomas are lethal brain tumors for which novel therapies are urgently needed. In animal models, vaccination with tumor-associated Ags efficiently primes T cells to clear gliomas. In clinical trials, cancer vaccines have been less effective at priming T cells and extending survival. Generalized immune suppression in the tumor draining lymph nodes has been documented in multiple cancers. However, a systematic analysis of how vaccination at various distances from the tumor (closest to farthest) has not been reported. We investigated how the injection site chosen for vaccination dictates CD8 T cell priming and survival in an OVA-transfected murine glioma model. Glioma-bearing mice were vaccinated with Poly:ICLC plus OVA protein in the neck, hind leg, or foreleg for drainage into the cervical, inguinal, or axillary lymph nodes, respectively. OVA-specific CD8 T cell number, TCR affinity, effector function, and infiltration into the brain decreased as the vaccination site approached the tumor. These effects were dependent on the presence of the tumor, because injection site did not appreciably affect CD8 T cell priming in tumor-free mice. Our data suggest the site of vaccination can greatly impact the effectiveness of cancer vaccines. Considering that previous and ongoing clinical trials have used a variety of injection sites, vaccination site is potentially a critical aspect of study design that is being overlooked.
Collapse
Affiliation(s)
- John R Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sage PT, Varghese LM, Martinelli R, Sciuto TE, Kamei M, Dvorak AM, Springer TA, Sharpe AH, Carman CV. Antigen recognition is facilitated by invadosome-like protrusions formed by memory/effector T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:3686-99. [PMID: 22442443 DOI: 10.4049/jimmunol.1102594] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adaptive immunity requires that T cells efficiently scan diverse cell surfaces to identify cognate Ag. However, the basic cellular mechanisms remain unclear. In this study, we investigated this process using vascular endothelial cells, APCs that possess a unique and extremely advantageous, planar morphology. High-resolution imaging revealed that CD4 memory/effector T cells dynamically probe the endothelium by extending submicron-scale, actin-rich "invadosome/podosome-like protrusions" (ILPs). The intimate intercellular contacts enforced by ILPs consistently preceded and supported T cell activation in response to endothelial MHC class II/Ag. The resulting calcium flux stabilized dense arrays of ILPs (each enriched in TCR, protein kinase C-θ, ZAP70, phosphotyrosine, and HS1), forming what we term a podo-synapse. Similar findings were made using CD8 CTLs on endothelium. Furthermore, careful re-examination of both traditional APC models and professional APCs suggests broad relevance for ILPs in facilitating Ag recognition. Together, our results indicate that ILPs function as sensory organelles that serve as actuators of immune surveillance.
Collapse
Affiliation(s)
- Peter T Sage
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Carrillo-de Sauvage MA, Gómez A, Ros CM, Ros-Bernal F, Martín ED, Perez-Vallés A, Gallego-Sanchez JM, Fernández-Villalba E, Barcia C, Barcia C, Herrero MT. CCL2-expressing astrocytes mediate the extravasation of T lymphocytes in the brain. Evidence from patients with glioma and experimental models in vivo. PLoS One 2012; 7:e30762. [PMID: 22319587 PMCID: PMC3271104 DOI: 10.1371/journal.pone.0030762] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 12/27/2011] [Indexed: 11/19/2022] Open
Abstract
CCL2 is a chemokine involved in brain inflammation, but the way in which it contributes to the entrance of lymphocytes in the parenchyma is unclear. Imaging of the cell type responsible for this task and details on how the process takes place in vivo remain elusive. Herein, we analyze the cell type that overexpresses CCL2 in multiple scenarios of T-cell infiltration in the brain and in three different species. We observe that CCL2+ astrocytes play a part in the infiltration of T-cells in the brain and our analysis shows that the contact of T-cells with perivascular astrocytes occurs, suggesting that may be an important event for lymphocyte extravasation.
Collapse
Affiliation(s)
- Maria Angeles Carrillo-de Sauvage
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
| | - Aurora Gómez
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
| | - Carmen María Ros
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
| | - Francisco Ros-Bernal
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
| | - Eduardo D. Martín
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), University of Castilla-La Mancha, Albacete, Spain
| | - Ana Perez-Vallés
- Department of Pathology, Hospital General Universitario de Valencia, Valencia, Spain
| | | | - Emiliano Fernández-Villalba
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
| | - Carlos Barcia
- Department of Neurosurgery, Hospital General Universitario de Valencia, Valencia, Spain
| | - Carlos Barcia
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
| | - Maria-Trinidad Herrero
- Clinical and Experimental Neuroscience, University of Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), School of Medicine, University of Murcia, Murcia, Spain
- * E-mail:
| |
Collapse
|
36
|
Salmon H, Franciszkiewicz K, Damotte D, Dieu-Nosjean MC, Validire P, Trautmann A, Mami-Chouaib F, Donnadieu E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest 2012; 122:899-910. [PMID: 22293174 DOI: 10.1172/jci45817] [Citation(s) in RCA: 753] [Impact Index Per Article: 57.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 12/14/2011] [Indexed: 01/01/2023] Open
Abstract
Appropriate localization and migration of T cells is a prerequisite for antitumor immune surveillance. Studies using fixed tumor samples from human patients have shown that T cells accumulate more efficiently in the stroma than in tumor islets, but the mechanisms by which this occurs are unknown. By combining immunostaining and real-time imaging in viable slices of human lung tumors, we revealed that the density and the orientation of the stromal extracellular matrix likely play key roles in controlling the migration of T cells. Active T cell motility, dependent on chemokines but not on β1 or β2 integrins, was observed in loose fibronectin and collagen regions, whereas T cells migrated poorly in dense matrix areas. Aligned fibers in perivascular regions and around tumor epithelial cell regions dictated the migratory trajectory of T cells and restricted them from entering tumor islets. Consistently, matrix reduction with collagenase increased the ability of T cells to contact cancer cells. Thus, the stromal extracellular matrix influences antitumor immunity by controlling the positioning and migration of T cells. Understanding the mechanisms by which this collagen network is generated has the potential to aid in the development of new therapeutics.
Collapse
Affiliation(s)
- Hélène Salmon
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Avril T, Vauleon E, Hamlat A, Saikali S, Etcheverry A, Delmas C, Diabira S, Mosser J, Quillien V. Human glioblastoma stem-like cells are more sensitive to allogeneic NK and T cell-mediated killing compared with serum-cultured glioblastoma cells. Brain Pathol 2011; 22:159-74. [PMID: 21790828 DOI: 10.1111/j.1750-3639.2011.00515.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most dramatic primary brain cancer with a very poor prognosis because of inevitable disease recurrence. The median overall survival is less than 1 year after diagnosis. Cancer stem cells have recently been disclosed in GBM. GBM stem-like cells (GSCs) exhibit resistance to radio/chemotherapeutic treatments and are therefore considered to play an important role in disease recurrence. GSCs are thus appealing targets for new treatments for GBM patients. In this study, we show that GBM cells with stem cell characteristics are resistant to lysis mediated by resting natural killer (NK) cells because of the expression of MHC class I molecules. However, GSCs are killed by lectin-activated NK cells. Furthermore, in experiments using the therapeutic antibody CetuximAb, we show that GSCs are sensitive to antibody-mediated cytotoxicity. We confirm the sensitivity of GSC to cytotoxicity carried out by IL2-activated NK cells and tumor-specific T cells. More importantly, we show that GSCs are more sensitive to NK and T cell-mediated lysis relatively to their corresponding serum-cultured GBM cells obtained from the same initial tumor specimen. Altogether, these results demonstrate the sensitivity of GSC to immune cell cytotoxicity and, therefore, strongly suggest that GSCs are suitable target cells for immunotherapy of GBM patients.
Collapse
Affiliation(s)
- Tony Avril
- Département de Biologie, Centre Eugène Marquis, Rennes, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wieërs G, Demotte N, Godelaine D, van der Bruggen P. Immune suppression in tumors as a surmountable obstacle to clinical efficacy of cancer vaccines. Cancers (Basel) 2011; 3:2904-54. [PMID: 24212939 PMCID: PMC3759179 DOI: 10.3390/cancers3032904] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/01/2011] [Accepted: 07/07/2011] [Indexed: 02/07/2023] Open
Abstract
Human tumors are usually not spontaneously eliminated by the immune system and therapeutic vaccination of cancer patients with defined antigens is followed by tumor regressions only in a small minority of the patients. The poor vaccination effectiveness could be explained by an immunosuppressive tumor microenvironment. Because T cells that infiltrate tumor metastases have an impaired ability to lyse target cells or to secrete cytokine, many researchers are trying to decipher the underlying immunosuppressive mechanisms. We will review these here, in particular those considered as potential therapeutic targets. A special attention will be given to galectins, a family of carbohydrate binding proteins. These lectins have often been implicated in inflammation and cancer and may be useful targets for the development of new anti-cancer therapies.
Collapse
Affiliation(s)
- Grégoire Wieërs
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Nathalie Demotte
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Danièle Godelaine
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| |
Collapse
|
39
|
Haque A, Banik NL, Ray SK. Molecular alterations in glioblastoma: potential targets for immunotherapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:187-234. [PMID: 21199773 DOI: 10.1016/b978-0-12-385506-0.00005-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma is the most common and deadly brain tumor, possibly arising from genetic and epigenetic alterations in normal astroglial cells. Multiple cytogenetic, chromosomal, and genetic alterations have been identified in glioblastoma, with distinct expression of antigens (Ags) and biomarkers that may alter therapeutic potential of this aggressive cancer. Current therapy consists of surgical resection, followed by radiation therapy and chemotherapy. In spite of these treatments, the prognosis for glioblastoma patients is poor. Although recent studies have focused on the development of novel immunotherapeutics against glioblastoma, little is known about glioblastoma-specific immune responses. A better understanding of the molecular interactions among glioblastoma tumors, host immune cells, and the tumor microenvironment may give rise to novel integrated approaches for the simultaneous control of tumor escape pathways and the activation of antitumor immune responses. This review provides a detailed overview concerning genetic alterations in glioblastoma, their effects on Ag and biomarker expression, and the future design of chemoimmunotherapeutics against glioblastoma.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | |
Collapse
|
40
|
Overview of cellular immunotherapy for patients with glioblastoma. Clin Dev Immunol 2010; 2010. [PMID: 20953324 PMCID: PMC2952949 DOI: 10.1155/2010/689171] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/13/2010] [Accepted: 08/27/2010] [Indexed: 12/22/2022]
Abstract
High grade gliomas (HGG) including glioblastomas (GBM) are the most common and devastating primary brain tumours. Despite important progresses in GBM treatment that currently includes surgery combined to radio- and chemotherapy, GBM patients' prognosis remains very poor. Immunotherapy is one of the new promising therapeutic approaches that can specifically target tumour cells. Such an approach could also maintain long term antitumour responses without inducing neurologic defects. Since the past 25 years, adoptive and active immunotherapies using lymphokine-activated killer cells, cytotoxic T cells, tumour-infiltrating lymphocytes, autologous tumour cells, and dendritic cells have been tested in phase I/II clinical trials with HGG patients. This paper inventories these cellular immunotherapeutic strategies and discusses their efficacy, limits, and future perspectives for optimizing the treatment to achieve clinical benefits for GBM patients.
Collapse
|
41
|
Yang J, Sanderson N, Wawrowsky K, Puntel M, Castro M, Lowenstein P. Kupfer-type immunological synapse characteristics do not predict anti-brain tumor cytolytic T-cell function in vivo. Proc Natl Acad Sci U S A 2010; 107:4716-21. [PMID: 20133734 PMCID: PMC2842057 DOI: 10.1073/pnas.0911587107] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To analyze the in vivo structure of antigen-specific immunological synapses during an effective immune response, we established brain tumors expressing the surrogate tumor antigen ovalbumin and labeled antigen-specific anti-glioma T cells using specific tetramers. Using these techniques, we determined that a significant number of antigen-specific T cells were localized to the brain tumor and surrounding brain tissue and a large percentage could be induced to express IFNgamma when exposed to the specific ovalbumin-derived peptide epitope SIINFEKL. Detailed morphological analysis of T cells immunoreactive for tetramers in direct physical contact with tumor cells expressing ovalbumin indicated that the interface between T cells and target tumor cells displayed various morphologies, including Kupfer-type immunological synapses. Quantitative analysis of adjacent confocal optical sections was performed to determine if the higher frequency of antigen-specific antiglioma T cells present in animals that developed an effective antitumor immune response could be correlated with a specific immunological synaptic morphology. Detailed in vivo quantitative analysis failed to detect an increased proportion of immunological synapses displaying the characteristic Kupfer-type morphology in animals mounting a strong and effective antitumor immune response as compared with those experiencing a clinically ineffective response. We conclude that an effective cytolytic immune response is not dependent on an increased frequency of Kupfer-type immunological synapses between T cells and tumor cells.
Collapse
Affiliation(s)
- J. Yang
- Board of Governors’ Gene Therapeutics Research Institute, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Departments of Medicine, and
- Molecular and Medical Pharmacology, and
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024
| | - N.S.R. Sanderson
- Board of Governors’ Gene Therapeutics Research Institute, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Departments of Medicine, and
- Molecular and Medical Pharmacology, and
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024
| | - K. Wawrowsky
- Board of Governors’ Gene Therapeutics Research Institute, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Departments of Medicine, and
- Molecular and Medical Pharmacology, and
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024
| | - M. Puntel
- Board of Governors’ Gene Therapeutics Research Institute, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - M.G. Castro
- Board of Governors’ Gene Therapeutics Research Institute, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Departments of Medicine, and
- Molecular and Medical Pharmacology, and
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024
| | - P.R. Lowenstein
- Board of Governors’ Gene Therapeutics Research Institute, Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Departments of Medicine, and
- Molecular and Medical Pharmacology, and
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024
| |
Collapse
|
42
|
Progress and prospects: Immunobiology of gene therapy for neurodegenerative disease: prospects and risks. Gene Ther 2010; 17:448-58. [PMID: 20147982 DOI: 10.1038/gt.2010.2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gene therapy for neurological, and in particular neurodegenerative, disease is now a reality. A number of early phase clinical trials have been completed and several are currently in progress. In view of this, it is critically important to evaluate the immunological risk associated with neurological gene therapy, which has clear implications for trial safety and efficacy. Moreover, it is imperative in particular to identify factors indicating potential high risk. In the light of recent advances in understanding immune regulation in the central nervous system (CNS) and with the continued development of new gene delivery vectors, this review critically assesses the current knowledge of immunobiology within the CNS in terms of likely immunological risk pertaining to viral vectors and gene therapy applications for neurodegenerative disease.
Collapse
|