1
|
Yi MH, Lee J, Moon S, So E, Bang G, Moon KS, Lee KH. Divergent Crosstalk Between Microglia and T Cells in Brain Cancers: Implications for Novel Therapeutic Strategies. Biomedicines 2025; 13:216. [PMID: 39857798 PMCID: PMC11763300 DOI: 10.3390/biomedicines13010216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Brain cancers represent a formidable oncological challenge characterized by their aggressive nature and resistance to conventional therapeutic interventions. The tumor microenvironment has emerged as a critical determinant of tumor progression and treatment efficacy. Within this complex ecosystem, microglia and macrophages play fundamental roles, forming intricate networks with peripheral immune cell populations, particularly T cells. The precise mechanisms underlying microglial interactions with T cells and their contributions to immunosuppression remain incompletely understood. Methods: This review comprehensively examines the complex cellular dialogue between microglia and T cells in two prominent brain malignancies: primary glioblastoma and secondary brain metastases. Results: Through a comprehensive review of the current scientific literature, we explore the nuanced mechanisms through which microglial-T cell interactions modulate tumor growth and immune responses. Conclusions: Our analysis seeks to unravel the cellular communication pathways that potentially underpin tumor progression, with the ultimate goal of illuminating novel therapeutic strategies for brain cancer intervention.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea; (M.-H.Y.)
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Jinkyung Lee
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Subin Moon
- Department of Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - EunA So
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Geonhyeok Bang
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea; (M.-H.Y.)
| | - Kyung-Sub Moon
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea;
| | - Kyung-Hwa Lee
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
- Department of Pathology, Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea
| |
Collapse
|
2
|
Cheng H, Zhao Y, Hou X, Ling F, Wang J, Wang Y, Cao Y. Unveiling the therapeutic prospects of IFNW1 and IFNA21: insights into glioma pathogenesis and clinical significance. Neurogenetics 2024; 25:337-350. [PMID: 38958838 DOI: 10.1007/s10048-024-00769-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Glioma, a type of brain tumor, poses significant challenges due to its heterogeneous nature and limited treatment options. Interferon-related genes (IRGs) have emerged as potential players in glioma pathogenesis, yet their expression patterns and clinical implications remain to be fully elucidated. We conducted a comprehensive analysis to investigate the expression patterns and functional enrichment of IRGs in glioma. This involved constructing protein-protein interaction networks, heatmap analysis, survival curve plotting, diagnostic and prognostic assessments, differential expression analysis across glioma subgroups, GSVA, immune infiltration analysis, and drug sensitivity analysis. Our analysis revealed distinct expression patterns and functional enrichment of IRGs in glioma. Notably, IFNW1 and IFNA21 were markedly downregulated in glioma tissues compared to normal tissues, and higher expression levels were associated with improved overall survival and disease-specific survival. Furthermore, these genes showed diagnostic capabilities in distinguishing glioma tissues from normal tissues and were significantly downregulated in higher-grade and more aggressive gliomas. Differential expression analysis across glioma subgroups highlighted the association of IFNW1 and IFNA21 expression with key pathways and biological processes, including metabolic reprogramming and immune regulation. Immune infiltration analysis revealed their influence on immune cell composition in the tumor microenvironment. Additionally, elevated expression levels were associated with increased resistance to chemotherapeutic agents. Our findings underscore the potential of IFNW1 and IFNA21 as diagnostic biomarkers and prognostic indicators in glioma. Their roles in modulating glioma progression, immune response, and drug sensitivity highlight their significance as potential therapeutic targets. These results contribute to a deeper understanding of glioma biology and may inform the development of personalized treatment strategies for glioma patients.
Collapse
Affiliation(s)
- Hong Cheng
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou University, No.136 Jiangyang Middle Road, Yangzhou, 225000, Jiangsu, China.
| | - Yingjie Zhao
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou University, No.136 Jiangyang Middle Road, Yangzhou, 225000, Jiangsu, China
- Cardiovascular Medicine, The Third People's Hospital of Danyang, Danyang, 212300, Jiangsu, China
| | - Xiaoli Hou
- Yangzhou Vocational University Medical College, Yangzhou, 225000, Jiangsu, China
| | - Fang Ling
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou University, No.136 Jiangyang Middle Road, Yangzhou, 225000, Jiangsu, China
- Otorhinolaryngology, The Third People's Hospital of Danyang, Danyang, 212300, Jiangsu, China
| | - Jing Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou University, No.136 Jiangyang Middle Road, Yangzhou, 225000, Jiangsu, China
- Medicine Section, The Third People's Hospital of Danyang, Danyang, 212300, Jiangsu, China
| | - Yixia Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou University, No.136 Jiangyang Middle Road, Yangzhou, 225000, Jiangsu, China
| | - Yasen Cao
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou University, No.136 Jiangyang Middle Road, Yangzhou, 225000, Jiangsu, China
| |
Collapse
|
3
|
de la Visitación N, Chen W, Krishnan J, Van Beusecum JP, Amarnath V, Hennen EM, Zhao S, Saleem M, Ao M, Dikalov SI, Dikalova AE, Harrison DG, Patrick DM. Immunoproteasomal Processing of IsoLG-Adducted Proteins Is Essential for Hypertension. Circ Res 2024; 134:1276-1291. [PMID: 38623763 PMCID: PMC11081850 DOI: 10.1161/circresaha.124.324068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/30/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Hypertension is characterized by CD8+ (cluster differentiation 8) T cell activation and infiltration into peripheral tissues. CD8+ T cell activation requires proteasomal processing of antigenic proteins. It has become clear that isoLG (isolevuglandin)-adduced peptides are antigenic in hypertension; however, IsoLGs inhibit the constitutive proteasome. We hypothesized that immunoproteasomal processing of isoLG-adducts is essential for CD8+ T cell activation and inflammation in hypertension. METHODS IsoLG adduct processing was studied in murine dendritic cells (DCs), endothelial cells (ECs), and B8 fibroblasts. The role of the proteasome and the immunoproteasome in Ang II (angiotensin II)-induced hypertension was studied in C57BL/6 mice treated with bortezomib or the immunoproteasome inhibitor PR-957 and by studying mice lacking 3 critical immunoproteasome subunits (triple knockout mouse). We also examined hypertension in mice lacking the critical immunoproteasome subunit LMP7 (large multifunctional peptidase 7) specifically in either DCs or ECs. RESULTS We found that oxidant stress increases the presence of isoLG adducts within MHC-I (class I major histocompatibility complex), and immunoproteasome overexpression augments this. Pharmacological or genetic inhibition of the immunoproteasome attenuated hypertension and tissue inflammation. Conditional deletion of LMP7 in either DCs or ECs attenuated hypertension and vascular inflammation. Finally, we defined the role of the innate immune receptors STING (stimulator of interferon genes) and TLR7/8 (toll-like receptor 7/8) as drivers of LMP7 expression in ECs. CONCLUSIONS These studies define a previously unknown role of the immunoproteasome in DCs and ECs in CD8+ T cell activation. The immunoproteasome in DCs and ECs is critical for isoLG-adduct presentation to CD8+ T cells, and in the endothelium, this guides homing and infiltration of T cells to specific tissues.
Collapse
Affiliation(s)
- Néstor de la Visitación
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wei Chen
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jaya Krishnan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin P. Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Charleston South Carolina
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Venkataraman Amarnath
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Shilin Zhao
- Vanderbilt Center for Quantitative Science, Vanderbilt University Medical Center
| | - Mohammad Saleem
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mingfang Ao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sergey I. Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anna E. Dikalova
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
- Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
4
|
He W, Wang N, Wang Y, Liu M, Qing Q, Su Q, Zou Y, Liu Y. Engineering Nanomedicine for Non-Viral RNA-Based Gene Therapy of Glioblastoma. Pharmaceutics 2024; 16:482. [PMID: 38675144 PMCID: PMC11054437 DOI: 10.3390/pharmaceutics16040482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of malignant tumor of the central nervous system, characterized by aggressiveness, genetic instability, heterogenesis, and unpredictable clinical behavior. Disappointing results from the current clinical therapeutic methods have fueled a search for new therapeutic targets and treatment modalities. GBM is characterized by various genetic alterations, and RNA-based gene therapy has raised particular attention in GBM therapy. Here, we review the recent advances in engineered non-viral nanocarriers for RNA drug delivery to treat GBM. Therapeutic strategies concerning the brain-targeted delivery of various RNA drugs involving siRNA, microRNA, mRNA, ASO, and short-length RNA and the therapeutical mechanisms of these drugs to tackle the challenges of chemo-/radiotherapy resistance, recurrence, and incurable stem cell-like tumor cells of GBM are herein outlined. We also highlight the progress, prospects, and remaining challenges of non-viral nanocarriers-mediated RNA-based gene therapy.
Collapse
Affiliation(s)
- Wenya He
- School of Pharmacy, Henan University, Kaifeng 475004, China; (W.H.)
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Ningyang Wang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Yaping Wang
- School of Pharmacy, Henan University, Kaifeng 475004, China; (W.H.)
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Mengyao Liu
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qian Qing
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qihang Su
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Yan Zou
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
- Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Yang Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng 475004, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China
| |
Collapse
|
5
|
Hiebinger F, Kudulyte A, Chi H, Burbano De Lara S, Ilic D, Helm B, Welsch H, Dao Thi VL, Klingmüller U, Binder M. Tumour cells can escape antiproliferative pressure by interferon-β through immunoediting of interferon receptor expression. Cancer Cell Int 2023; 23:315. [PMID: 38066598 PMCID: PMC10709914 DOI: 10.1186/s12935-023-03150-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2025] Open
Abstract
Type I interferons (IFNs) play a central role not only in innate immunity against viral infection, but also in the antitumour response, e.g. through a direct impact on cell proliferation. Particularly for cancer arising in the context of chronic inflammation, constant exposure to IFNs may constitute a strong selective pressure during tumour evolution. Expansion of neoplastic subclones resistant to the antiproliferative effects of IFNs may contribute to immunoediting of tumours, leading to more aggressive disease. Experimental evidence for this development of IFN-insensitivity has been scarce and its molecular mechanism is unclear. In this study we demonstrate that six weeks exposure of cells to IFN-β in vitro reduces their sensitivity to its antiproliferative effects, and that this phenotype was stable for up to four weeks. Furthermore, we observed substantial differences in cellular sensitivity to growth inhibition by IFN-β in a panel of ten different liver cancer cell lines, most prominently in a pair of highly dedifferentiated cell lines, and least in cells from well-differentiated tumours. In both, long-term IFN selection and in dedifferentiated tumour cell lines, we found IFNAR2 expression to be substantially reduced, suggesting the receptor complex to be a sensitive target amenable to immunoediting. Beyond new insights into possible molecular processes in tumour evolution, these findings might prove valuable for the development of biomarkers allowing to stratify tumours for their sensitivity to IFN treatment in the context of patient tailored therapies.
Collapse
Affiliation(s)
- Felix Hiebinger
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Aiste Kudulyte
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Huanting Chi
- Schaller Research Group, Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Sebastian Burbano De Lara
- Division of Systems Biology of Signal Transduction (B200), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Doroteja Ilic
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Barbara Helm
- Division of Systems Biology of Signal Transduction (B200), German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Lung Research (DZL) and Translational Lung Research Center Heidelberg (TRLC), Heidelberg, Germany
| | - Hendrik Welsch
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Viet Loan Dao Thi
- Schaller Research Group, Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Ursula Klingmüller
- Division of Systems Biology of Signal Transduction (B200), German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Lung Research (DZL) and Translational Lung Research Center Heidelberg (TRLC), Heidelberg, Germany
| | - Marco Binder
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
6
|
Castillo SP, Galvez-Cancino F, Liu J, Pollard SM, Quezada SA, Yuan Y. The tumour ecology of quiescence: Niches across scales of complexity. Semin Cancer Biol 2023; 92:139-149. [PMID: 37037400 DOI: 10.1016/j.semcancer.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/06/2023] [Accepted: 04/08/2023] [Indexed: 04/12/2023]
Abstract
Quiescence is a state of cell cycle arrest, allowing cancer cells to evade anti-proliferative cancer therapies. Quiescent cancer stem cells are thought to be responsible for treatment resistance in glioblastoma, an aggressive brain cancer with poor patient outcomes. However, the regulation of quiescence in glioblastoma cells involves a myriad of intrinsic and extrinsic mechanisms that are not fully understood. In this review, we synthesise the literature on quiescence regulatory mechanisms in the context of glioblastoma and propose an ecological perspective to stemness-like phenotypes anchored to the contemporary concepts of niche theory. From this perspective, the cell cycle regulation is multiscale and multidimensional, where the niche dimensions extend to extrinsic variables in the tumour microenvironment that shape cell fate. Within this conceptual framework and powered by ecological niche modelling, the discovery of microenvironmental variables related to hypoxia and mechanosignalling that modulate proliferative plasticity and intratumor immune activity may open new avenues for therapeutic targeting of emerging biological vulnerabilities in glioblastoma.
Collapse
Affiliation(s)
- Simon P Castillo
- Centre for Evolution and Cancer & Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Felipe Galvez-Cancino
- Immune Regulation and Tumor Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Jiali Liu
- Immune Regulation and Tumor Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine and Cancer Research UK Scotland Centre, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sergio A Quezada
- Immune Regulation and Tumor Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer & Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK.
| |
Collapse
|
7
|
de la Visitación N, Chen W, Krishnan J, Van Beusecum JP, Amarnath V, Hennen EM, Zhao S, Saleem M, Ao M, Harrison DG, Patrick DM. Immunoproteasomal Processing of Isolevuglandin Adducts in Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536054. [PMID: 37383945 PMCID: PMC10299468 DOI: 10.1101/2023.04.10.536054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Isolevuglandins (isoLGs) are lipid aldehydes that form in the presence of reactive oxygen species (ROS) and drive immune activation. We found that isoLG-adducts are presented within the context of major histocompatibility complexes (MHC-I) by an immunoproteasome dependent mechanism. Pharmacologic inhibition of LMP7, the chymotrypsin subunit of the immunoproteasome, attenuates hypertension and tissue inflammation in the angiotensin II (Ang II) model of hypertension. Genetic loss of function of all immunoproteasome subunits or conditional deletion of LMP7 in dendritic cell (DCs) or endothelial cells (ECs) attenuated hypertension, reduced aortic T cell infiltration, and reduced isoLG-adduct MHC-I interaction. Furthermore, isoLG adducts structurally resemble double-stranded DNA and contribute to the activation of STING in ECs. These studies define a critical role of the immunoproteasome in the processing and presentation of isoLG-adducts. Moreover they define a role of LMP7 as a regulator of T cell activation and tissue infiltration in hypertension.
Collapse
|
8
|
Kong Y, Xue Z, Wang H, Cui G, Chen A, Liu J, Wang J, Li X, Huang B. Identification of BST2 Contributing to the Development of Glioblastoma Based on Bioinformatics Analysis. Front Genet 2022; 13:890174. [PMID: 35865015 PMCID: PMC9294273 DOI: 10.3389/fgene.2022.890174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/15/2022] [Indexed: 11/22/2022] Open
Abstract
Rigorous molecular analysis of the immune cell environment and immune response of human tumors has led to immune checkpoint inhibitors as one of the most promising strategies for the treatment of human cancer. However, in human glioblastoma multiforme (GBM) which develops in part by attracting immune cell types intrinsic to the human brain (microglia), standard immunotherapy has yielded inconsistent results in experimental models and patients. Here, we analyzed publicly available expression datasets to identify molecules possibly associated with immune response originating from or influencing the tumor microenvironment in primary tumor samples. Using three glioma datasets (GSE16011, Rembrandt-glioma and TCGA-glioma), we first analyzed the data to distinguish between GBMs of high and low tumor cell purity, a reflection of the cellular composition of the tumor microenvironment, and second, to identify differentially expressed genes (DEGs) between these two groups using GSEA and other analyses. Tumor purity was negatively correlated with patient prognosis. The interferon gamma-related gene BST2 emerged as a DEG that was highly expressed in GBM and negatively correlated with tumor purity. BST2high tumors also tended to harbor PTEN mutations (31 vs. 9%, BST2high versus BST2low) while BST2low tumors more often had sustained TP53 mutations (8 versus 36%, BST2high versus BST2low). Prognosis of patients with BST2high tumors was also poor relative to patients with BST2low tumors. Further molecular in silico analysis demonstrated that high expression of BST2 was negatively correlated with CD8+ T cells but positively correlated with macrophages with an M2 phenotype. Further functional analysis demonstrated that BST2 was associated with multiple immune checkpoints and cytokines, and may promote tumorigenesis and progression through interferon gamma, IL6/JAK/STAT3 signaling, IL2/STAT5 signaling and the TNF-α signaling via NF-kB pathway. Finally, a series of experiments confirmed that the expression of BST2 can be significantly increased by IFN induction, and knockdown of BST2 can significantly inhibit the growth and invasion of GBM cells, and may affect the phenotype of tumor-associated macrophages. In conclusion, BST2 may promote the progression of GBM and may be a target for treatment.
Collapse
Affiliation(s)
- Yang Kong
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Neurological Care Unit, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Haiying Wang
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Guangqiang Cui
- Neurological Care Unit, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jie Liu
- Neurological Care Unit, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- *Correspondence: Bin Huang,
| |
Collapse
|
9
|
Yan D, Li W, Liu Q, Yang K. Advances in Immune Microenvironment and Immunotherapy of Isocitrate Dehydrogenase Mutated Glioma. Front Immunol 2022; 13:914618. [PMID: 35769466 PMCID: PMC9234270 DOI: 10.3389/fimmu.2022.914618] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022] Open
Abstract
The tumor immune microenvironment and immunotherapy have become current important tumor research concerns. The unique immune microenvironment plays a crucial role in the malignant progression of isocitrate dehydrogenase (IDH) mutant gliomas. IDH mutations in glioma can inhibit tumor-associated immune system evasion of NK cell immune surveillance. Meanwhile, mutant IDH can inhibit classical and alternative complement pathways and directly inhibit T-cell responses by metabolizing isocitrate to D-2-Hydroxyglutaric acid (2-HG). IDH has shown clinically relevant efficacy as a potential target for immunotherapy. This article intends to summarize the research progress in the immunosuppressive microenvironment and immunotherapy of IDH-mutant glioma in recent years in an attempt to provide new ideas for the study of occurrence, progression, and treatment of IDH-mutant glioma.
Collapse
Affiliation(s)
- Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Weicheng Li
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qibing Liu
- Department of Pharmacology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Qibing Liu, ; Kun Yang,
| | - Kun Yang
- Department of Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Qibing Liu, ; Kun Yang,
| |
Collapse
|
10
|
Cattolico C, Bailey P, Barry ST. Modulation of Type I Interferon Responses to Influence Tumor-Immune Cross Talk in PDAC. Front Cell Dev Biol 2022; 10:816517. [PMID: 35273962 PMCID: PMC8902310 DOI: 10.3389/fcell.2022.816517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy has revolutionized the treatment of many cancer types. However, pancreatic ductal adenocarcinomas (PDACs) exhibit poor responses to immune checkpoint inhibitors with immunotherapy-based trials not generating convincing clinical activity. PDAC tumors often have low infiltration of tumor CD8+ T cells and a highly immunosuppressive microenvironment. These features classify PDAC as immunologically "cold." However, the presence of tumor T cells is a favorable prognostic feature in PDAC. Intrinsic tumor cell properties govern interactions with the immune system. Alterations in tumor DNA such as genomic instability, high tumor mutation burden, and/or defects in DNA damage repair are associated with responses to both immunotherapy and chemotherapy. Cytotoxic or metabolic stress produced by radiation and/or chemotherapy can act as potent immune triggers and prime immune responses. Damage- or stress-mediated activation of nucleic acid-sensing pathways triggers type I interferon (IFN-I) responses that activate innate immune cells and natural killer cells, promote maturation of dendritic cells, and stimulate adaptive immunity. While PDAC exhibits intrinsic features that have the potential to engage immune cells, particularly following chemotherapy, these immune-sensing mechanisms are ineffective. Understanding where defects in innate immune triggers render the PDAC tumor-immune interface less effective, or how T-cell function is suppressed will help develop more effective treatments and harness the immune system for durable outcomes. This review will focus on the pivotal role played by IFN-I in promoting tumor cell-immune cell cross talk in PDAC. We will discuss how PDAC tumor cells bypass IFN-I signaling pathways and explore how these pathways can be co-opted or re-engaged to enhance the therapeutic outcome.
Collapse
Affiliation(s)
- Carlotta Cattolico
- Bioscience, Early Oncology, AstraZeneca, Cambridge, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Peter Bailey
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
- Section Surgical Research, University Clinic Heidelberg, Heidelberg, Germany
| | - Simon T. Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
11
|
SARS-CoV-2 inhibits induction of the MHC class I pathway by targeting the STAT1-IRF1-NLRC5 axis. Nat Commun 2021; 12:6602. [PMID: 34782627 PMCID: PMC8594428 DOI: 10.1038/s41467-021-26910-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
The MHC class I-mediated antigen presentation pathway plays a critical role in antiviral immunity. Here we show that the MHC class I pathway is targeted by SARS-CoV-2. Analysis of the gene expression profile from COVID-19 patients as well as SARS-CoV-2 infected epithelial cell lines reveals that the induction of the MHC class I pathway is inhibited by SARS-CoV-2 infection. We show that NLRC5, an MHC class I transactivator, is suppressed both transcriptionally and functionally by the SARS-CoV-2 ORF6 protein, providing a mechanistic link. SARS-CoV-2 ORF6 hampers type II interferon-mediated STAT1 signaling, resulting in diminished upregulation of NLRC5 and IRF1 gene expression. Moreover, SARS-CoV-2 ORF6 inhibits NLRC5 function via blocking karyopherin complex-dependent nuclear import of NLRC5. Collectively, our study uncovers an immune evasion mechanism of SARS-CoV-2 that targets the function of key MHC class I transcriptional regulators, STAT1-IRF1-NLRC5. The presentation of viral antigens to T cells via the MHC molecules is a critical component of the host response to viral infection. Here the authors suggest SARS-CoV-2 possesses the immune evasion strategy against the MHC class I pathway by targeting key transcriptional regulators.
Collapse
|
12
|
Xiao Z, Zhang W, Li G, Li W, Li L, Sun T, He Y, Liu G, Wang L, Han X, Wen H, Liu Y, Chen Y, Wang H, Li J, Fan Y, Zhang J. Multiomics Analysis Reveals the Prognostic Non-tumor Cell Landscape in Glioblastoma Niches. Front Genet 2021; 12:741325. [PMID: 34603399 PMCID: PMC8481948 DOI: 10.3389/fgene.2021.741325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/11/2021] [Indexed: 11/21/2022] Open
Abstract
A comprehensive characterization of non-tumor cells in the niches of primary glioblastoma is not fully established yet. This study aims to present an overview of non-malignant cells in the complex microenvironment of glioblastoma with detailed characterizations of their prognostic effects. We curate 540 gene signatures covering a total of 64 non-tumor cell types. Cell type-specific expression patterns are interrogated by normalized enrichment score across four large gene expression profiling cohorts of glioblastoma with a total number of 967 cases. The glioblastoma multiforms (GBMs) in each cohort are hierarchically clustered into negative or positive immune response classes with significantly different overall survival. Our results show that astrocytes, macrophages, monocytes, NKTs, and MSC are risk factors, while CD8 T cells, CD8 naive T cells, and plasma cells are protective factors. Moreover, we find that the immune system and organogenesis are uniformly enriched in negative immune response clusters, in contrast to the enrichment of nervous system in positive immune response clusters. Mesenchymal differentiation is also observed in the negative immune response clusters. High enrichment status of macrophages in negative immune response clusters is independently validated by analyzing scRNA-seq data from eight high-grade gliomas, revealing that negative immune response samples comprised 46.63 to 55.12% of macrophages, whereas positive immune response samples comprised only 1.70 to 8.12%, with IHC staining of samples from six short-term and six long-term survivors of GBMs confirming the results.
Collapse
Affiliation(s)
- Zixuan Xiao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wei Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wendong Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lin Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Ting Sun
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yufei He
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Guang Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xiaohan Han
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Hao Wen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yong Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yifan Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Haoyu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
13
|
Ji H, Ba Y, Ma S, Hou K, Mi S, Gao X, Jin J, Gong Q, Liu T, Wang F, Liu Z, Li S, Du J, Hu S. Construction of Interferon-Gamma-Related Gene Signature to Characterize the Immune-Inflamed Phenotype of Glioblastoma and Predict Prognosis, Efficacy of Immunotherapy and Radiotherapy. Front Immunol 2021; 12:729359. [PMID: 34566988 PMCID: PMC8461254 DOI: 10.3389/fimmu.2021.729359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Interferon-gamma (IFNG) has profound impacts on tumor-immune interaction and is of great clinical significance for multiple cancers. Exploring the role of IFNG in glioblastoma (GBM) may optimize the current treatment paradigm of this disease. Here, multi-dimensional data of 429 GBM samples were collected. Various bioinformatics algorithms were employed to establish a gene signature that characterizes immunological features, genomic alterations, and clinical characteristics associated with the IFNG response. In this way, a novel IFNG-related gene signature (IFNGrGS, including TGFBI, IL4I1, ACP5, and LUM) has been constructed and validated. Samples with increased IFNGrGS scores were characterized by increased neutrophil and macrophage infiltration and exuberant innate immune responses, while the activated adaptive immune response may be frustrated by multiple immunosuppressive mechanisms. Notably, the IFNG pathway as well as its antagonistic pathways including IL4, IL10, TGF-beta, and VEGF converged on the expression of immune checkpoints. Besides, gene mutations involved in the microenvironment were associated with the IFNGrGS-based stratification, where the heterogeneous prognostic significance of EGFR mutation may be related to the different degrees of IFNG response. Moreover, the IFNGrGS score had solid prognostic value and the potential to screen ICB and radiotherapy sensitive populations. Collectively, our study provided insights into the role of IFNG on the GBM immune microenvironment and offered feasible information for optimizing the treatment of GBM.
Collapse
Affiliation(s)
- Hang Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yixu Ba
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Shuai Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Kuiyuan Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Shan Mi
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xin Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaqi Jin
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Qin Gong
- School of Life Sciences, Nanjing University, Nanjing, China
| | - Ting Liu
- Faculty of Pharmacy, Harbin Medical University (DAQING), Daqing, China
| | - Fang Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Zhihui Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Shupeng Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianyang Du
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shaoshan Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Neurosurgery, Emergency Medicine Center, Zhejiang Provincial People's Hospital Affiliated to Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
14
|
Ricon-Becker I, Fogel E, Cole SW, Haldar R, Lev-Ari S, Gidron Y. Tone it down: Vagal nerve activity is associated with pro-inflammatory and anti-viral factors in breast cancer – An exploratory study. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2021; 7:100057. [PMID: 35757058 PMCID: PMC9216392 DOI: 10.1016/j.cpnec.2021.100057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/28/2022] Open
Abstract
In response to adverse social-environmental conditions, leukocytes gene expression profile is altered in a pattern recognized as the conserved transcriptional response to adversity (CTRA). This entails the up-regulated expression of pro-inflammatory genes and down-regulated expression of genes involved in type-I interferon (IFN) related anti-viral immunity. In contrast, vagal nerve activity is recognized as a significant anti-inflammatory modulator. In this work, we investigated the association between CTRA and vagal activity indicated by the standard deviation of all NN interval (SDNN), a measure of heart-rate variability, in breast cancer patients awaiting surgery (n = 16). This association was tested both at the molecular leukocyte transcription factor activity level, as well as at the cytokines serum levels. We found an association between higher SDNN and increased interferon (IFN) related anti-viral pathways, both on the leukocyte transcription factor level and serum protein level. Unexpectedly, we also found a positive correlation between higher SDNN and pro-inflammatory transcription factor activity and cytokine serum level, potentially suggesting that increased vagal activity was induced by increased inflammation, in the context of pre-surgical stress and the presence of malignant tissue. Transcription origin analysis (TOA) suggests a role for monocyte and B-cells in the anti-inflammatory and anti-metastatic effects induced by vagal nerve signaling. Larger prospective studies are needed to verify and elaborate on the results from this small cross-sectional study. Increased HRV is associated with increased anti-viral immunity. Unexpectedly, HRV is also associated with increased pro-inflammatory signaling. Findings are evident separately at transcription factor activity level as well as at cytokine serum levels. Macrophages and B cells emerge as the origin of these differences. Findings may shed light on novel pathways in which vagal nerve activity modulate cancer progression.
Collapse
Affiliation(s)
- Itay Ricon-Becker
- School of Psychological Sciences, Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
- Corresponding author. Neuroimmunology Research Unit, Prof. Shamgar Ben-Eliyahu's Laboratory, School of Psychological Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| | - Efrat Fogel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Steve W. Cole
- Division of Hematology-Oncology, Department of Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, USA
| | - Rita Haldar
- School of Psychological Sciences, Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Shahar Lev-Ari
- Department of Health Promotion, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Yori Gidron
- Faculty of Health Sciences and Welfare University of Haifa, Haifa, Israel
| |
Collapse
|
15
|
Huot N, Rascle P, Petitdemange C, Contreras V, Stürzel CM, Baquero E, Harper JL, Passaes C, Legendre R, Varet H, Madec Y, Sauermann U, Stahl-Hennig C, Nattermann J, Saez-Cirion A, Le Grand R, Keith Reeves R, Paiardini M, Kirchhoff F, Jacquelin B, Müller-Trutwin M. SIV-induced terminally differentiated adaptive NK cells in lymph nodes associated with enhanced MHC-E restricted activity. Nat Commun 2021; 12:1282. [PMID: 33627642 PMCID: PMC7904927 DOI: 10.1038/s41467-021-21402-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells play a critical understudied role during HIV infection in tissues. In a natural host of SIV, the African green monkey (AGM), NK cells mediate a strong control of SIVagm infection in secondary lymphoid tissues. We demonstrate that SIVagm infection induces the expansion of terminally differentiated NKG2alow NK cells in secondary lymphoid organs displaying an adaptive transcriptional profile and increased MHC-E-restricted cytotoxicity in response to SIV Env peptides while expressing little IFN-γ. Such NK cell differentiation was lacking in SIVmac-infected macaques. Adaptive NK cells displayed no increased NKG2C expression. This study reveals a previously unknown profile of NK cell adaptation to a viral infection, thus accelerating strategies toward NK-cell directed therapies and viral control in tissues.
Collapse
Affiliation(s)
- Nicolas Huot
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | - Philippe Rascle
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France ,grid.508487.60000 0004 7885 7602Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Caroline Petitdemange
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | - Vanessa Contreras
- CEA-Université Paris Sud-Inserm, U1184, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | | | - Eduard Baquero
- grid.462718.eInstitut Pasteur, Unité de Virologie Structurale, Paris, France
| | - Justin L. Harper
- grid.189967.80000 0001 0941 6502Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA USA
| | - Caroline Passaes
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | - Rachel Legendre
- grid.428999.70000 0001 2353 6535Bioinformatics and Biostatistics Hub, Department of Computational Biology, Institut Pasteur, Paris, France
| | - Hugo Varet
- grid.428999.70000 0001 2353 6535Biomics Platform, Center for Technological Resources and Research (C2RT), Institut Pasteur, Paris, France
| | - Yoann Madec
- grid.428999.70000 0001 2353 6535 Institut Pasteur; Epidemiology of Emerging Diseases Unit, Paris, France
| | - Ulrike Sauermann
- grid.418215.b0000 0000 8502 7018Deutsches Primatenzentrum - Leibniz Institut für Primatenforschung, Göttingen, Germany
| | - Christiane Stahl-Hennig
- grid.418215.b0000 0000 8502 7018Deutsches Primatenzentrum - Leibniz Institut für Primatenforschung, Göttingen, Germany
| | - Jacob Nattermann
- grid.452463.2Medizinische Klinik und Poliklinik I, Universitätsklinikum Bonn, Germany; German Center for Infection Research (DZIF), Bonn, Germany
| | - Asier Saez-Cirion
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | - Roger Le Grand
- CEA-Université Paris Sud-Inserm, U1184, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - R. Keith Reeves
- grid.38142.3c000000041936754XCenter for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Mirko Paiardini
- grid.189967.80000 0001 0941 6502Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA USA
| | | | - Beatrice Jacquelin
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | - Michaela Müller-Trutwin
- grid.428999.70000 0001 2353 6535Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| |
Collapse
|
16
|
Zou SS, Qiao Y, Zhu S, Gao B, Yang N, Liu YJ, Chen J. Intrinsic strategies for the evasion of cGAS-STING signaling-mediated immune surveillance in human cancer: How therapy can overcome them. Pharmacol Res 2021; 166:105514. [PMID: 33631336 DOI: 10.1016/j.phrs.2021.105514] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS) recognizes cytosolic DNA and catalyzes the formation of cyclic GMP-AMP, which upon activation triggers the induction of stimulator of interferon genes (STING), leading to type I interferons production; these events then promote the cross-priming of dendritic cells and the initiation of a tumor-specific CD8+ T cell response. However, cancer cells in the tumor microenvironment cannot trigger intrinsic cGAS-STING signaling, regardless of the expression of cGAS and STING. This dysfunctional cGAS-STING signaling enables cancer cells to evade immune surveillance, thereby promoting tumorigenesis. Here, we review recent advances in the current understanding of the activation of cGAS-STING signaling and immunotherapies based on this pathway and focus on the mechanisms for the inactivation of this pathway in tumor cells to promote the development of cancer immunotherapy. The discovery of inherent resistance and the selection of appropriate combination therapies are of great significance for tumor treatment development.
Collapse
Affiliation(s)
- Shan-Shan Zou
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yuan Qiao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Bao Gao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ning Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
17
|
MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12071760. [PMID: 32630675 PMCID: PMC7409324 DOI: 10.3390/cancers12071760] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, major advances have been made in cancer immunotherapy. This has led to significant improvement in prognosis of cancer patients, especially in the hematological setting. Nonetheless, translation of these successes to solid tumors was found difficult. One major mechanism through which solid tumors can avoid anti-tumor immunity is the downregulation of major histocompatibility complex class I (MHC-I), which causes reduced recognition by- and cytotoxicity of CD8+ T-cells. Downregulation of MHC-I has been described in 40-90% of human tumors, often correlating with worse prognosis. Epigenetic and (post-)transcriptional dysregulations relevant in the stabilization of NFkB, IRFs, and NLRC5 are often responsible for MHC-I downregulation in cancer. The intrinsic reversible nature of these dysregulations provides an opportunity to restore MHC-I expression and facilitate adaptive anti-tumor immunity. In this review, we provide an overview of the mechanisms underlying reversible MHC-I downregulation and describe potential strategies to counteract this reduction in MHC-I antigen presentation in cancer.
Collapse
|
18
|
Rajani KR, Carlstrom LP, Parney IF, Johnson AJ, Warrington AE, Burns TC. Harnessing Radiation Biology to Augment Immunotherapy for Glioblastoma. Front Oncol 2019; 8:656. [PMID: 30854331 PMCID: PMC6395389 DOI: 10.3389/fonc.2018.00656] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most common adult primary brain tumor and carries a dismal prognosis. Radiation is a standard first-line therapy, typically deployed following maximal safe surgical debulking, when possible, in combination with cytotoxic chemotherapy. For other systemic cancers, standard of care is being transformed by immunotherapies, including checkpoint-blocking antibodies targeting CTLA-4 and PD-1/PD-L1, with potential for long-term remission. Ongoing studies are evaluating the role of immunotherapies for GBM. Despite dramatic responses in some cases, randomized trials to date have not met primary outcomes. Challenges have been attributed in part to the immunologically "cold" nature of glioblastoma relative to other malignancies successfully treated with immunotherapy. Radiation may serve as a mechanism to improve tumor immunogenicity. In this review, we critically evaluate current evidence regarding radiation as a synergistic facilitator of immunotherapies through modulation of both the innate and adaptive immune milieu. Although current preclinical data encourage efforts to harness synergistic biology between radiation and immunotherapy, several practical and scientific challenges remain. Moreover, insights from radiation biology may unveil additional novel opportunities to help mobilize immunity against GBM.
Collapse
Affiliation(s)
- Karishma R. Rajani
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Lucas P. Carlstrom
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Ian F. Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Terry C. Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
19
|
van Vloten JP, Santry LA, McAusland TM, Karimi K, McFadden G, Petrik JJ, Wootton SK, Bridle BW. Quantifying Antigen-Specific T Cell Responses When Using Antigen-Agnostic Immunotherapies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 13:154-166. [PMID: 30788384 PMCID: PMC6369252 DOI: 10.1016/j.omtm.2019.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 01/16/2019] [Indexed: 12/21/2022]
Abstract
Immunotherapies are at the forefront of the fight against cancers, and researchers continue to develop and test novel immunotherapeutic modalities. Ideal cancer immunotherapies induce a patient’s immune system to kill their own cancer and develop long-lasting immunity. Research has demonstrated a critical requirement for CD8+ and CD4+ T cells in achieving durable responses. In the path to the clinic, researchers require robust tools to effectively evaluate the capacity for immunotherapies to generate adaptive anti-tumor responses. To study functional tumor-specific T cells, researchers have relied on targeting tumor-associated antigens (TAAs) or the inclusion of surrogate transgenes in pre-clinical models, which facilitate detection of T cells by using the targeted antigen(s) in peptide re-stimulation or tetramer-staining assays. Unfortunately, many pre-clinical models lack a defined TAA, and epitope mapping of TAAs is costly. Surrogate transgenes can alter tumor engraftment and influence the immunogenicity of tumors, making them less relevant to clinical tumors. Further, some researchers prefer to develop therapies that do not rely on pre-defined TAAs. Here, we describe a method to exploit major histocompatibility complex expression on murine cancer cell lines in a co-culture assay to detect T cells responding to bulk, undefined, tumor antigens. This is a tool to support the preclinical evaluation of novel, antigen-agnostic immunotherapies.
Collapse
Affiliation(s)
- Jacob P van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Lisa A Santry
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Thomas M McAusland
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Grant McFadden
- The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - James J Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Sarah K Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
20
|
Medrano RF, Hunger A, Mendonça SA, Barbuto JAM, Strauss BE. Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy. Oncotarget 2017; 8:71249-71284. [PMID: 29050360 PMCID: PMC5642635 DOI: 10.18632/oncotarget.19531] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
During the last decades, the pleiotropic antitumor functions exerted by type I interferons (IFNs) have become universally acknowledged, especially their role in mediating interactions between the tumor and the immune system. Indeed, type I IFNs are now appreciated as a critical component of dendritic cell (DC) driven T cell responses to cancer. Here we focus on IFN-α and IFN-β, and their antitumor effects, impact on immune responses and their use as therapeutic agents. IFN-α/β share many properties, including activation of the JAK-STAT signaling pathway and induction of a variety of cellular phenotypes. For example, type I IFNs drive not only the high maturation status of DCs, but also have a direct impact in cytotoxic T lymphocytes, NK cell activation, induction of tumor cell death and inhibition of angiogenesis. A variety of stimuli, including some standard cancer treatments, promote the expression of endogenous IFN-α/β, which then participates as a fundamental component of immunogenic cell death. Systemic treatment with recombinant protein has been used for the treatment of melanoma. The induction of endogenous IFN-α/β has been tested, including stimulation through pattern recognition receptors. Gene therapies involving IFN-α/β have also been described. Thus, harnessing type I IFNs as an effective tool for cancer therapy continues to be studied.
Collapse
Affiliation(s)
- Ruan F.V. Medrano
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aline Hunger
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Samir Andrade Mendonça
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| | - José Alexandre M. Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Cell and Molecular Therapy Center, NUCEL-NETCEM, University of São Paulo, São Paulo, Brazil
| | - Bryan E. Strauss
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM 24, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
21
|
Nguyen T, Lagman C, Chung LK, Chen CHJ, Poon J, Ong V, Voth BL, Yang I. Insights into CCL21's roles in immunosurveillance and immunotherapy for gliomas. J Neuroimmunol 2017; 305:29-34. [PMID: 28284342 DOI: 10.1016/j.jneuroim.2017.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/17/2017] [Indexed: 02/02/2023]
Abstract
Chemokine (C-C) motif ligand 21 (CCL21) is involved in immunosurveillance and has recently garnered the attention of neuro-oncologists and neuroscientists. CCL21 contains an extended C-terminus, which increases binding to lymphatic glycosaminoglycans and provides a mechanism for cell trafficking by forming a stationary chemokine concentration gradient that allows cell migration via haptotaxis. CCL21 is expressed by endothelial cells of the blood-brain barrier in physiologic and pathologic conditions. CCL21 has also been implicated in leukocyte extravasation into the central nervous system. In this review, we summarize the role of CCL21 in immunosurveillance and explore its potential as an immunotherapeutic agent for the treatment of gliomas.
Collapse
Affiliation(s)
- Thien Nguyen
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Carlito Lagman
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lawrance K Chung
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cheng Hao Jacky Chen
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jessica Poon
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Vera Ong
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Brittany L Voth
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isaac Yang
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States; Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, CA, United States; Department of Head and Neck Surgery, University of California, Los Angeles, Los Angeles, CA, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
22
|
Tan MP, Dolton GM, Gerry AB, Brewer JE, Bennett AD, Pumphrey NJ, Jakobsen BK, Sewell AK. Human leucocyte antigen class I-redirected anti-tumour CD4 + T cells require a higher T cell receptor binding affinity for optimal activity than CD8 + T cells. Clin Exp Immunol 2016; 187:124-137. [PMID: 27324616 PMCID: PMC5167017 DOI: 10.1111/cei.12828] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2016] [Indexed: 12/12/2022] Open
Abstract
CD4+ T helper cells are a valuable component of the immune response towards cancer. Unfortunately, natural tumour‐specific CD4+ T cells occur in low frequency, express relatively low‐affinity T cell receptors (TCRs) and show poor reactivity towards cognate antigen. In addition, the lack of human leucocyte antigen (HLA) class II expression on most cancers dictates that these cells are often unable to respond to tumour cells directly. These deficiencies can be overcome by transducing primary CD4+ T cells with tumour‐specific HLA class I‐restricted TCRs prior to adoptive transfer. The lack of help from the co‐receptor CD8 glycoprotein in CD4+ cells might result in these cells requiring a different optimal TCR binding affinity. Here we compared primary CD4+ and CD8+ T cells expressing wild‐type and a range of affinity‐enhanced TCRs specific for the HLA A*0201‐restricted NY‐ESO‐1‐ and gp100 tumour antigens. Our major findings are: (i) redirected primary CD4+ T cells expressing TCRs of sufficiently high affinity exhibit a wide range of effector functions, including cytotoxicity, in response to cognate peptide; and (ii) optimal TCR binding affinity is higher in CD4+ T cells than CD8+ T cells. These results indicate that the CD4+ T cell component of current adoptive therapies using TCRs optimized for CD8+ T cells is below par and that there is room for substantial improvement.
Collapse
Affiliation(s)
- M P Tan
- Cardiff University School of Medicine, Cardiff, UK
| | - G M Dolton
- Cardiff University School of Medicine, Cardiff, UK
| | | | | | | | | | | | - A K Sewell
- Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
23
|
Mostafa H, Pala A, Högel J, Hlavac M, Dietrich E, Westhoff MA, Nonnenmacher L, Burster T, Georgieff M, Wirtz CR, Schneider EM. Immune phenotypes predict survival in patients with glioblastoma multiforme. J Hematol Oncol 2016; 9:77. [PMID: 27585656 PMCID: PMC5009501 DOI: 10.1186/s13045-016-0272-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/13/2016] [Indexed: 11/24/2022] Open
Abstract
Background Glioblastoma multiforme (GBM), a common primary malignant brain tumor, rarely disseminates beyond the central nervous system and has a very bad prognosis. The current study aimed at the analysis of immunological control in individual patients with GBM. Methods Immune phenotypes and plasma biomarkers of GBM patients were determined at the time of diagnosis using flow cytometry and ELISA, respectively. Results Using descriptive statistics, we found that immune anomalies were distinct in individual patients. Defined marker profiles proved highly relevant for survival. A remarkable relation between activated NK cells and improved survival in GBM patients was in contrast to increased CD39 and IL-10 in patients with a detrimental course and very short survival. Recursive partitioning analysis (RPA) and Cox proportional hazards models substantiated the relevance of absolute numbers of CD8 cells and low numbers of CD39 cells for better survival. Conclusions Defined alterations of the immune system may guide the course of disease in patients with GBM and may be prognostically valuable for longitudinal studies or can be applied for immune intervention. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0272-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haouraa Mostafa
- Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.,Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - Andrej Pala
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Josef Högel
- Institute for Human Genetics, Albert Einstein Allee 11, 89081, Ulm, Germany
| | - Michal Hlavac
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Elvira Dietrich
- Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.,Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - M Andrew Westhoff
- Department of Pediatric Hematology and Oncology, University Hospital Ulm, Prittwitzstr. 43, 89075, Ulm, Germany
| | - Lisa Nonnenmacher
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Timo Burster
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - Michael Georgieff
- Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany
| | - C Rainer Wirtz
- Department of Neurosurgery, University Hospital Ulm Albert Einstein Allee 23, 89081 Ulm and Bezirkskrankenhaus Günzburg, Ludwig-Heilmeyer-Str. 2, 89312, Günzburg, Germany
| | - E Marion Schneider
- Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany. .,Klinik für Anaesthesiologie, University Hospital Ulm, Albert Einstein Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
24
|
Chawla A, Alatrash G, Philips AV, Qiao N, Sukhumalchandra P, Kerros C, Diaconu I, Gall V, Neal S, Peters HL, Clise-Dwyer K, Molldrem JJ, Mittendorf EA. Neutrophil elastase enhances antigen presentation by upregulating human leukocyte antigen class I expression on tumor cells. Cancer Immunol Immunother 2016; 65:741-51. [PMID: 27129972 PMCID: PMC5764112 DOI: 10.1007/s00262-016-1841-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 04/09/2016] [Indexed: 12/24/2022]
Abstract
Neutrophil elastase (NE) is an innate immune cell-derived inflammatory mediator that we have shown increases the presentation of tumor-associated peptide antigens in breast cancer. In this study, we extend these observations to show that NE uptake has a broad effect on enhancing antigen presentation by breast cancer cells. We show that NE increases human leukocyte antigen (HLA) class I expression on the surface of breast cancer cells in a concentration and time-dependent manner. HLA class I upregulation requires internalization of enzymatically active NE. Western blots of NE-treated breast cancer cells confirm that the expression of total HLA class I as well as the antigen-processing machinery proteins TAP1, LMP2, and calnexin does not change following NE treatment. This suggests that NE does not increase the efficiency of antigen processing; rather, it mediates the upregulation of HLA class I by stabilizing and reducing membrane recycling of HLA class I molecules. Furthermore, the effects of NE extend beyond breast cancer since the uptake of NE by EBV-LCL increases the presentation of HLA class I-restricted viral peptides, as shown by their increased sensitivity to lysis by EBV-specific CD8+ T cells. Together, our results show that NE uptake increases the responsiveness of breast cancer cells to adaptive immunity by broad upregulation of membrane HLA class I and support the conclusion that the innate inflammatory mediator NE enhances tumor cell recognition and increases tumor sensitivity to the host adaptive immune response.
Collapse
Affiliation(s)
- Akhil Chawla
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gheath Alatrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0900, Houston, TX, 77030, USA.
| | - Anne V Philips
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Na Qiao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pariya Sukhumalchandra
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0900, Houston, TX, 77030, USA
| | - Celine Kerros
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0900, Houston, TX, 77030, USA
| | - Iulia Diaconu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Victor Gall
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samantha Neal
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haley L Peters
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0900, Houston, TX, 77030, USA
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0900, Houston, TX, 77030, USA
| | - Jeffrey J Molldrem
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0900, Houston, TX, 77030, USA
| | - Elizabeth A Mittendorf
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1434, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Modeling the Treatment of Glioblastoma Multiforme and Cancer Stem Cells with Ordinary Differential Equations. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2016; 2016:1239861. [PMID: 27022405 PMCID: PMC4745194 DOI: 10.1155/2016/1239861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/12/2015] [Accepted: 12/22/2015] [Indexed: 11/24/2022]
Abstract
Despite improvements in cancer therapy and treatments, tumor recurrence is a common event in cancer patients. One explanation of recurrence is that cancer therapy focuses on treatment of tumor cells and does not eradicate cancer stem cells (CSCs). CSCs are postulated to behave similar to normal stem cells in that their role is to maintain homeostasis. That is, when the population of tumor cells is reduced or depleted by treatment, CSCs will repopulate the tumor, causing recurrence. In this paper, we study the application of the CSC Hypothesis to the treatment of glioblastoma multiforme by immunotherapy. We extend the work of Kogan et al. (2008) to incorporate the dynamics of CSCs, prove the existence of a recurrence state, and provide an analysis of possible cancerous states and their dependence on treatment levels.
Collapse
|
26
|
Abstract
The interferons (IFNs) are a family of cytokines that protect against disease by direct effects on target cells and by activating immune responses. The production and actions of IFNs are finely tuned to achieve maximal protection and avoid the potential toxicity associated with excessive responses. IFNs are back in the spotlight owing to mounting evidence that is reshaping how we can exploit this pathway therapeutically. As IFNs can be produced by, and act on, both tumour cells and immune cells, understanding this reciprocal interaction will enable the development of improved single-agent or combination therapies that exploit IFN pathways and new 'omics'-based biomarkers to indicate responsive patients.
Collapse
Affiliation(s)
- Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Jai Rautela
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Ayari C, Besançon M, Bergeron A, LaRue H, Bussières V, Fradet Y. Poly(I:C) potentiates Bacillus Calmette-Guérin immunotherapy for bladder cancer. Cancer Immunol Immunother 2016; 65:223-34. [PMID: 26759009 PMCID: PMC11029542 DOI: 10.1007/s00262-015-1789-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 12/27/2015] [Indexed: 02/04/2023]
Abstract
Non-specific immunotherapy consisting of intravesical instillation of Bacillus Calmette-Guérin (BCG) is currently the best available treatment to prevent non-muscle-invasive bladder tumor recurrence and progression. This treatment however is suboptimal, and more effective immunotherapeutic approaches are needed. Toll-like receptors (TLRs) play a major role in the activation of the immune system in response to pathogens and danger signals but also in anti-tumor responses. We previously showed that human urothelial cells express functional TLRs and respond to TLR2 and TLR3 agonists. In this study, we analyzed the potential of polyinosinic:polycytidylic acid [poly(I:C)], a TLR3 agonist, to replace or complement BCG in the treatment of non-muscle-invasive bladder cancer. We observed that poly(I:C) had an anti-proliferative, cytotoxic, and apoptotic effect in vitro on two low-grade human bladder cancer cell lines, MGH-U3 and RT4. In MGH-U3 cells, poly(I:C) induced growth arrest at the G1-S transition. Poly(I:C) also increased the immunogenicity of MGH-U3 and RT4 cells, inducing the secretion of MHC class I molecules and of pro-inflammatory cytokines. By comparison, poly(I:C) had less in vitro impact on two high-grade human bladder cancer cell lines, 5637 and T24, and on MBT-2 murine high-grade bladder cancer cells. The latter can be used as an immunocompetent model of bladder cancer. The combination poly(I:C)/BCG was much more effective in reducing MBT-2 tumor growth in mice than either treatment alone. It completely cured 29% of mice and also induced an immunological memory response. In conclusion, our study suggests that adding poly(I:C) to BCG may enhance the therapeutic effect of BCG.
Collapse
Affiliation(s)
- Cherifa Ayari
- Centre de recherche sur le cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval, L'Hôtel-Dieu de Québec, 10 McMahon, Québec, G1R 3S1, Canada
| | - Marjorie Besançon
- Centre de recherche sur le cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval, L'Hôtel-Dieu de Québec, 10 McMahon, Québec, G1R 3S1, Canada
| | - Alain Bergeron
- Centre de recherche sur le cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval, L'Hôtel-Dieu de Québec, 10 McMahon, Québec, G1R 3S1, Canada
| | - Hélène LaRue
- Centre de recherche sur le cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval, L'Hôtel-Dieu de Québec, 10 McMahon, Québec, G1R 3S1, Canada.
| | - Vanessa Bussières
- Centre de recherche sur le cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval, L'Hôtel-Dieu de Québec, 10 McMahon, Québec, G1R 3S1, Canada
| | - Yves Fradet
- Centre de recherche sur le cancer de l'Université Laval, Centre de recherche du CHU de Québec-Université Laval, L'Hôtel-Dieu de Québec, 10 McMahon, Québec, G1R 3S1, Canada
| |
Collapse
|
28
|
Burks J, Reed RE, Desai SD. Free ISG15 triggers an antitumor immune response against breast cancer: a new perspective. Oncotarget 2016; 6:7221-31. [PMID: 25749047 PMCID: PMC4466680 DOI: 10.18632/oncotarget.3372] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
Interferon-Stimulated Gene 15 (ISG15), an antagonist of the canonical ubiquitin pathway, is frequently overexpressed in various cancers. In cancer cells, ISG15 is detected as free (intracellular) and conjugated to cellular proteins (ISGylation). Free ISG15 is also secreted into the extracellular milieu. ISGylation has protumor functions and extracellular free ISG15 has immunomodulatory properties in vitro. Therefore, whether ISG15 is a tumor suppressor or tumor promoter in vivo remains controversial. The current study aimed to clarify the role of free ISG15 in tumorigenesis. Breast cancer cells stably expressing control, ISG15, and UbcH8 (ISG15-specific E2 ligase) shRNAs were used to assess the immunoregulatory and antitumor function of free ISG15 in cell culture (in vitro) and in nude mice (in vivo). We show that extracellular free ISG15 suppresses breast tumor growth and increases NK cell infiltration into xenografted breast tumors in nude mice, and intracellular free ISG15 enhances major histocompatibility complex (MHC) class I surface expression in breast cancer cells. We conclude that free ISG15 may have antitumor and immunoregulatory function in vivo. These findings provides the basis for developing strategies to increase systemic levels of free ISG15 to treat cancer patients overexpressing the ISG15 pathway.
Collapse
Affiliation(s)
- Julian Burks
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, New Orleans, LA, USA.,Present Address: Georgetown University Medical Center, Lombardi Comprehensive Cancer Center Department of Molecular Oncology, Washington, DC, USA
| | - Ryan E Reed
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, New Orleans, LA, USA
| | - Shyamal D Desai
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, New Orleans, LA, USA
| |
Collapse
|
29
|
Dobreva A, Paus R, Cogan N. Mathematical model for alopecia areata. J Theor Biol 2015; 380:332-45. [DOI: 10.1016/j.jtbi.2015.05.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 05/14/2015] [Accepted: 05/21/2015] [Indexed: 11/16/2022]
|
30
|
Biomarkers for glioma immunotherapy: the next generation. J Neurooncol 2015; 123:359-72. [PMID: 25724916 DOI: 10.1007/s11060-015-1746-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/16/2015] [Indexed: 12/11/2022]
Abstract
The term "biomarker" historically refers to a single parameter, such as the expression level of a gene or a radiographic pattern, used to indicate a broader biological state. Molecular indicators have been applied to several aspects of cancer therapy: to describe the genotypic and phenotypic state of neoplastic tissue for prognosis, to predict susceptibility to anti-proliferative agents, to validate the presence of specific drug targets, and to evaluate responsiveness to therapy. For glioblastoma (GBM), immunohistochemical and radiographic biomarkers accessible to the clinical lab have informed traditional regimens, but while immunotherapies have emerged as potentially disruptive weapons against this diffusely infiltrating, heterogeneous tumor, biomarkers with strong predictive power have not been fully established. The cancer immunotherapy field, through the recently accelerated expansion of trials, is currently leveraging this wealth of clinical and biological data to define and revise the use of biomarkers for improving prognostic accuracy, personalization of therapy, and evaluation of responses across the wide variety of tumors. Technological advancements in DNA sequencing, cytometry, and microscopy have facilitated the exploration of more integrated, high-dimensional profiling of the disease system-incorporating both immune and tumor parameters-rather than single metrics, as biomarkers for therapeutic sensitivity. Here we discuss the utility of traditional GBM biomarkers in immunotherapy and how the impending transformation of the biomarker paradigm-from single markers to integrated profiles-may offer the key to bringing predictive, personalized immunotherapy to GBM patients.
Collapse
|
31
|
Angell TE, Lechner MG, Jang JK, LoPresti JS, Epstein AL. MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in vitro. Clin Cancer Res 2014; 20:6034-44. [PMID: 25294906 DOI: 10.1158/1078-0432.ccr-14-0879] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE To evaluate MHC class I expression on papillary thyroid cancer (PTC) and analyze changes in MHC expression and associated immune activation with current and experimental treatments for thyroid cancer using in vitro PTC cell lines. EXPERIMENTAL DESIGN MHC class I expression and assessment of tumor-infiltrating leukocyte populations were evaluated by immunohistochemistry. PTC cell lines were analyzed for HLA-ABC expression by flow cytometry following tyrosine kinase inhibitor, IFNα or IFNγ, or radiation treatment. Functional changes in antigenicity were assessed by coculture of allogeneic donor peripheral blood leukocytes (PBL) with pretreated or untreated PTC cell lines and measurement of T-cell activation and cytokine production. RESULTS Both MHC class I and β2-microglobulin expression was reduced or absent in 76% of PTC specimens and was associated with reduced tumor-infiltrating immune cells, including effector (CD3(+), CD8(+), CD16(+)) and suppressor (FoxP3(+)) populations. Treatment of PTC cell lines with the MEK1/2 inhibitor selumetinib or IFN increased HLA-ABC expression. This phenotypic change was associated with increased T-cell activation (%CD25(+) of CD3(+)) and IL2 production by PBL cocultured with treated PTC cell lines. Additive effects were seen with combination selumetinib and IFN treatment. CONCLUSIONS MHC class I expression loss is frequent in human PTC specimens and represents a significant mechanism of immune escape. Increased antigenicity following selumetinib and IFN treatment warrants further study for immunotherapy of progressive PTC.
Collapse
Affiliation(s)
- Trevor E Angell
- Division of Endocrinology and Diabetes, Keck Medical Center, University of Southern California, Los Angeles, California. Department of Pathology, Keck Medical Center, University of Southern California, Los Angeles, California. Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Melissa G Lechner
- Department of Pathology, Keck Medical Center, University of Southern California, Los Angeles, California. Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie K Jang
- Department of Pathology, Keck Medical Center, University of Southern California, Los Angeles, California
| | - Jonathan S LoPresti
- Division of Endocrinology and Diabetes, Keck Medical Center, University of Southern California, Los Angeles, California
| | - Alan L Epstein
- Department of Pathology, Keck Medical Center, University of Southern California, Los Angeles, California.
| |
Collapse
|
32
|
Swartz AM, Li QJ, Sampson JH. Rindopepimut: a promising immunotherapeutic for the treatment of glioblastoma multiforme. Immunotherapy 2014; 6:679-90. [PMID: 25186601 PMCID: PMC4524671 DOI: 10.2217/imt.14.21] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive glial cell-derived primary tumor. Current standard of care for patients with GBM includes maximal tumor resection plus adjuvant radiotherapy and temozolomide chemotherapy, increasing median overall survival to a mere 15 months from diagnosis. Because these therapies are inherently nonspecific, there is an increased likelihood of off-target and incomplete effects; therefore, targeted modalities are required for enhanced safety and efficacy. Rindopepimut is emerging as a safe and potentially effective drug for the treatment of GBM. Rindopepimut consists of a 14-mer peptide that spans the length of EGF receptor variant III, a mutant variant of EGF receptor found on approximately 30% of primary GBM, conjugated to the carrier protein keyhole limpet hemocyanin. Vaccination with rindopepimut has been shown to specifically eliminate cells expressing EGF receptor variant III. Phase II clinical trials have suggested that vaccination of newly diagnosed GBM patients with rindopepimut plus adjuvant granulocyte-macrophage colony-stimulating factor results in prolonged progression-free and overall survival with minimal toxicity. This review will outline the development of rindopepimut, as well as the current status of this vaccine.
Collapse
Affiliation(s)
- AM Swartz
- Duke University Medical Center, Department of Surgery, Division of Neurosurgery, DUMC Box 3050, Durham, NC 27710, Phone: (919) 684-9041, Fax: (919) 684-9045
| | - QJ Li
- Duke University Medical Center, Department of Surgery, Division of Neurosurgery, DUMC Box 3050, Durham, NC 27710, Phone: (919) 684-9041, Fax: (919) 684-9045
| | - JH Sampson
- Duke University Medical Center, Department of Surgery, Division of Neurosurgery, DUMC Box 3050, Durham, NC 27710, Phone: (919) 684-9041, Fax: (919) 684-9045
| |
Collapse
|
33
|
Yeung JT, Hamilton RL, Okada H, Jakacki RI, Pollack IF. Increased expression of tumor-associated antigens in pediatric and adult ependymomas: implication for vaccine therapy. J Neurooncol 2013; 111:103-11. [PMID: 23179498 PMCID: PMC3546121 DOI: 10.1007/s11060-012-0998-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 10/31/2012] [Indexed: 12/28/2022]
Abstract
Despite surgery and radiotherapy, as many as 50 % of children with ependymomas will suffer from tumor recurrences that will ultimately lead to death. Our group's initial peptide-based glioma vaccine targeting EphA2, IL-13Rα2, and Survivin, which are overexpressed in pediatric gliomas, has shown promise in its initial phase of testing. We therefore investigated whether EphA2, IL-13Rα2, Survivin, and, additionally, Wilms' Tumor 1 (WT1), are overexpressed in pediatric ependymomas to determine if a similar immunotherapy approach could be applicable. Immunohistochemistry was performed using antibodies specific for EphA2, IL-13Rα2, Survivin, and WT1 on paraffin-embedded specimens from 19 pediatric and 13 adult ependymomas. Normal brain and ependyma were used for background staining controls. Negative staining was defined as no staining or staining equaling the background intensity in normal brain tissues. In the 19 pediatric cases, 18 (95 %) demonstrated positive staining for EphA2, 16 (84 %) for IL-13Rα2, 18 (95 %) for Survivin, and only 7 (37 %) for WT1. Only 3 of 19 cases were positive for two or fewer tumor-associated antigens (TAAs); 16 of 19 cases were positive for three or more TAAs. In the 13 adult cases, all 13 demonstrated positive staining for EphA2, IL-13Rα2, and Survivin. Only 2 of 13 cases (15 %) demonstrated positive staining for WT1. All adult specimens were positive for three or more TAAs. Some ependymomas showed patchy variability in intensity. Pediatric and adult ependymomas frequently express EphA2, IL-13Rα2, and Survivin. This provides the basis for the utilization of an established multiple peptide vaccine for ependymoma in a clinical trial setting.
Collapse
Affiliation(s)
- Jacky T. Yeung
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ronald L. Hamilton
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Regina I. Jakacki
- Departments of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Ian F. Pollack
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Children’s Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA,
| |
Collapse
|
34
|
Zolotukhin I, Luo D, Gorbatyuk O, Hoffman B, Warrington K, Herzog R, Harrison J, Cao O. Improved Adeno-associated Viral Gene Transfer to Murine Glioma. ACTA ACUST UNITED AC 2013; 4. [PMID: 24319629 DOI: 10.4172/2157-7412.1000133] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Glioblastoma (GBM) is a deadly primary brain tumor. Current treatment, consisting of surgical removal of the tumor mass followed by chemotherapy and/or radiotherapy, does not significantly prolong survival. Gene therapies for GBM are being developed in clinical trials, for example using adenoviral vectors. While adeno-associated virus (AAV) represents an alternative vector system, limited gene transfer to glioma cells has hampered its use. Here, we evaluated newly emerged variants of AAV capsid for gene delivery to murine glioma. We tested a mutant AAV2 capsid devoid of 3 surface-exposed tyrosine residues, AAV2 (Y444-500-730F), and a "shuffed" capsid (ShH19, containing sequences from several serotypes) that had previously been selected for enhanced glial gene delivery. AAV2 (Y-F) and ShH19 showed improved transduction of murine glioma GL261 cells in vitro by 2- to 6-fold, respectively, over AAV2. While AAV2 gene transfer to GL261 cells in established tumors in brains of syngeneic mice was undetectable, intratumoral injection of AAV2 (Y-F) or ShH19 resulted in local transduction of approximately 10% of tumor cells. In addition, gene transfer to neurons adjacent to the tumor was observed, while microglia were rarely transduced. Use of self-complementary vectors further increased transduction of glioma cells. Together, the data demonstrate the potential for improved AAV-based gene therapy for glioma using recently developed capsid variants.
Collapse
Affiliation(s)
- I Zolotukhin
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Nieto-Sampedro M, Valle-Argos B, Gómez-Nicola D, Fernández-Mayoralas A, Nieto-Díaz M. Inhibitors of Glioma Growth that Reveal the Tumour to the Immune System. Clin Med Insights Oncol 2011; 5:265-314. [PMID: 22084619 PMCID: PMC3201112 DOI: 10.4137/cmo.s7685] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Treated glioblastoma patients survive from 6 to 14 months. In the first part of this review, we describe glioma origins, cancer stem cells and the genomic alterations that generate dysregulated cell division, with enhanced proliferation and diverse response to radiation and chemotherapy. We review the pathways that mediate tumour cell proliferation, neo-angiogenesis, tumor cell invasion, as well as necrotic and apoptotic cell death. Then, we examine the ability of gliomas to evade and suppress the host immune system, exhibited at the levels of antigen recognition and immune activation, limiting the effective signaling between glioma and host immune cells.The second part of the review presents current therapies and their drawbacks. This is followed by a summary of the work of our laboratory during the past 20 years, on oligosaccharide and glycosphingolipid inhibitors of astroblast and astrocytoma division. Neurostatins, the O-acetylated forms of gangliosides GD1b and GT1b naturally present in mammalian brain, are cytostatic for normal astroblasts, but cytotoxic for rat C6 glioma cells and human astrocytoma grades III and IV, with ID50 values ranging from 200 to 450 nM. The inhibitors do not affect neurons or fibroblasts up to concentrations of 4 μM or higher.At least four different neurostatin-activated, cell-mediated antitumoral processes, lead to tumor destruction: (i) inhibition of tumor neovascularization; (ii) activation of microglia; (iii) activation of natural killer (NK) cells; (iv) activation of cytotoxic lymphocytes (CTL). The enhanced antigenicity of neurostatin-treated glioma cells, could be related to their increased expression of connexin 43. Because neurostatins and their analogues show specific activity and no toxicity for normal cells, a clinical trial would be the logical next step.
Collapse
Affiliation(s)
- Manuel Nieto-Sampedro
- Instituto Cajal de Neurobiología, CSIC, 28002 Madrid, Spain
- Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Beatriz Valle-Argos
- Instituto Cajal de Neurobiología, CSIC, 28002 Madrid, Spain
- Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Diego Gómez-Nicola
- Instituto Cajal de Neurobiología, CSIC, 28002 Madrid, Spain
- Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | | | | |
Collapse
|
37
|
Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AKM, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2011; 11:155-80. [PMID: 21453286 DOI: 10.2174/156652311795684722] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Maria G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yang I, Han SJ, Sughrue ME, Tihan T, Parsa AT. Immune cell infiltrate differences in pilocytic astrocytoma and glioblastoma: evidence of distinct immunological microenvironments that reflect tumor biology. J Neurosurg 2011; 115:505-11. [PMID: 21663411 DOI: 10.3171/2011.4.jns101172] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECT The tumor microenvironment in astrocytomas is composed of a variety of cell types, including infiltrative inflammatory cells that are dynamic in nature, potentially reflecting tumor biology. In this paper the authors demonstrate that characterization of the intratumoral inflammatory infiltrate can distinguish high-grade glioblastoma from low-grade pilocytic astrocytoma. METHODS Tumor specimens from ninety-one patients with either glioblastoma or pilocytic astrocytoma were analyzed at the University of California, San Francisco. A systematic neuropathology analysis was performed. All tissue was collected at the time of the initial surgery prior to adjuvant treatment. Immune cell infiltrate not associated with necrosis or hemorrhage was analyzed on serial 4-μm sections. Analysis was performed for 10 consecutive hpfs and in 3 separate regions (total 30 × 0.237 mm(2)). Using immunohistochemistry for markers of infiltrating cytotoxic T cells (CD8), natural killer cells (CD56), and macrophages (CD68), the inflammatory infiltrates in these tumors were graded quantitatively and classified based on microanatomical location (perivascular vs intratumoral). Control markers included CD3, CD20, and human leukocyte antigen. RESULTS Glioblastomas exhibited significantly higher perivascular (CD8) T-cell infiltration than pilocytic astrocytomas (62% vs 29%, p = 0.0005). Perivascular (49%) and intratumoral (89%; p = 0.004) CD56-positive cells were more commonly associated with glioblastoma. The CD68-positive cells also were more prevalent in the perivascular and intratumoral space in glioblastoma. In the intratumoral space, all glioblastomas exhibited CD68-positive cells compared with 86% of pilocytic astrocytomas (p = 0.0014). Perivascularly, CD68-positive infiltrate was also more prevalent in glioblastoma when compared with pilocytic astrocytoma (97% vs 86%, respectively; p = 0.0003). The CD3-positive, CD20-positive, and human leukocyte antigen-positive infiltrates did not differ between glioblastoma and pilocytic astrocytoma. CONCLUSIONS This analysis suggests a significantly distinct immune profile in the microenvironment of high-grade glioblastoma versus low-grade pilocytic astrocytoma. This difference in tumor microenvironment may reflect an important difference in the tumor biology of glioblastoma.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurosurgery, University of California, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
39
|
Prins RM, Soto H, Konkankit V, Odesa SK, Eskin A, Yong WH, Nelson SF, Liau LM. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res 2010; 17:1603-15. [PMID: 21135147 DOI: 10.1158/1078-0432.ccr-10-2563] [Citation(s) in RCA: 330] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE To assess the feasibility, safety, and toxicity of autologous tumor lysate-pulsed dendritic cell (DC) vaccination and toll-like receptor (TLR) agonists in patients with newly diagnosed and recurrent glioblastoma. Clinical and immune responses were monitored and correlated with tumor gene expression profiles. EXPERIMENTAL DESIGN Twenty-three patients with glioblastoma (WHO grade IV) were enrolled in this dose-escalation study and received three biweekly injections of glioma lysate-pulsed DCs followed by booster vaccinations with either imiquimod or poly-ICLC adjuvant every 3 months until tumor progression. Gene expression profiling, immunohistochemistry, FACS, and cytokine bead arrays were performed on patient tumors and peripheral blood mononuclear cells. RESULTS DC vaccinations are safe and not associated with any dose-limiting toxicity. The median overall survival from the time of initial surgical diagnosis of glioblastoma was 31.4 months, with a 1-, 2-, and 3-year survival rate of 91%, 55%, and 47%, respectively. Patients whose tumors had mesenchymal gene expression signatures exhibited increased survival following DC vaccination compared with historic controls of the same genetic subtype. Tumor samples with a mesenchymal gene expression signature had a higher number of CD3(+) and CD8(+) tumor-infiltrating lymphocytes compared with glioblastomas of other gene expression signatures (P = 0.006). CONCLUSION Autologous tumor lysate-pulsed DC vaccination in conjunction with TLR agonists is safe as adjuvant therapy in newly diagnosed and recurrent glioblastoma patients. Our results suggest that the mesenchymal gene expression profile may identify an immunogenic subgroup of glioblastoma that may be more responsive to immune-based therapies.
Collapse
Affiliation(s)
- Robert M Prins
- Department of Neurosurgery, Brain Research Institute, The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California 90095-6901, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Iwahashi M, Katsuda M, Nakamori M, Nakamura M, Naka T, Ojima T, Iida T, Yamaue H. Vaccination with peptides derived from cancer-testis antigens in combination with CpG-7909 elicits strong specific CD8+ T cell response in patients with metastatic esophageal squamous cell carcinoma. Cancer Sci 2010; 101:2510-7. [PMID: 20874827 PMCID: PMC11158191 DOI: 10.1111/j.1349-7006.2010.01732.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Potent helper action is necessary for peptide-based vaccines to efficiently induce antitumor immune responses against advanced cancer. A phase I trial for advanced esophageal squamous cell carcinoma was carried out for patients with HLA-A*2402 using epitope peptides derived from novel cancer-testis antigens, LY6K and TTK, in combination with CpG-7909 (NCT00669292). This study investigated the feasibility and the toxicity as well as induction of tumor antigen-specific immune responses. Nine patients were vaccinated on days 1, 8, 15, and 22 of each 28-day treatment cycle with peptide LY6K-177, peptide TTK-567, and CpG-7909 (level-1; 0, level-2; 0.02, level-3; 0.1 mg/kg) and all were tolerated by this treatment. LY6K-specific T cell responses in PBMCs were detected in two of the three patients in each level. In particular, two patients in level-2/3 showed potent LY6K-specific T cell responses. In contrast, only two patients in level-2/3 showed TTK-567-specific T cell responses. The frequency of LY6K-177 or TTK-567-specific CD8+ T cells increased in patients in level-2/3 (with CpG). The vaccination with peptides and CpG-7909 increased and activated both plasmacytoid dendritic cells and natural killer cells, and increased the serum level of α-interferon. There were no complete response (CR) and partial response (PR), however, one of three patients in level-1, and four of six patients in level-2/3 showed stable disease (SD). In conclusion, vaccination with LY6K-177 and TTK-567 in combination with CpG-7909 successfully elicited antigen-specific CD8+ T cell responses and enhanced the innate immunity of patients with advanced esophageal squamous cell carcinoma. This vaccine protocol is therefore recommended to undergo further phase II trials.
Collapse
Affiliation(s)
- Makoto Iwahashi
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sigalov AB. The SCHOOL of nature: IV. Learning from viruses. SELF/NONSELF 2010; 1:282-298. [PMID: 21487503 PMCID: PMC3062383 DOI: 10.4161/self.1.4.13279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/04/2010] [Accepted: 08/05/2010] [Indexed: 02/05/2023]
Abstract
During the co-evolution of viruses and their hosts, the latter have equipped themselves with an elaborate immune system to defend themselves from the invading viruses. In order to establish a successful infection, replicate and persist in the host, viruses have evolved numerous strategies to counter and evade host antiviral immune responses as well as exploit them for productive viral replication. These strategies include those that modulate signaling mediated by cell surface receptors. Despite tremendous advancement in recent years, the exact molecular mechanisms underlying these critical points in viral pathogenesis remain unknown. In this work, based on a novel platform of receptor signaling, the Signaling Chain HOmoOLigomerization (SCHOOL) platform, I suggest specific mechanisms used by different viruses such as human immunodeficiency virus (HIV), cytomegalovirus (CMV), severe acute respiratory syndrome coronavirus, human herpesvirus 6 and others, to modulate receptor signaling. I also use the example of HIV and CMV to illustrate how two unrelated enveloped viruses use a similar SCHOOL mechanism to modulate the host immune response mediated by two functionally different receptors: T cell antigen receptor and natural killer cell receptor, NKp30. This suggests that it is very likely that similar general mechanisms can be or are used by other viral and possibly non-viral pathogens. Learning from viruses how to target cell surface receptors not only helps us understand viral strategies to escape from the host immune surveillance, but also provides novel avenues in rational drug design and the development of new therapies for immune disorders.
Collapse
|
42
|
Waldron JS, Yang I, Han S, Tihan T, Sughrue ME, Mills SA, Pieper RO, Parsa AT. Implications for immunotherapy of tumor-mediated T-cell apoptosis associated with loss of the tumor suppressor PTEN in glioblastoma. J Clin Neurosci 2010; 17:1543-7. [PMID: 20822910 DOI: 10.1016/j.jocn.2010.04.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 04/22/2010] [Accepted: 04/25/2010] [Indexed: 12/31/2022]
Abstract
The ability of glioma cells to escape the immune system remains a significant barrier to successful immunotherapy. Here we demonstrate that loss of the PTEN tumor suppressor gene, with associated activation of the PI3K/Akt/mTOR pathway, leads to a human glioma phenotype that induces autologous T-cell apoptosis upon contact. The PTEN status of pathologically confirmed glioblastoma specimens was defined, and primary cultures established after surgical resection of tumor from 26 patients. Autologous T-cells were isolated from these patients, and after T-cell activation was induced, these cells were co-cultured with matched autologous glioma cells, either alone, or after treatment with one of three inhibitors of the PI3K/Akt/mTOR pathway. When co-cultured with autologous T-cells, PTEN wild-type tumor cells induced apoptosis in a minimal number of activated T-cells (6-12% of T-cells), whereas tumors with PTEN loss induced much more profound levels of T-cell apoptosis (42-56% of T-cells). Prior treatment of PTEN-deficient tumor cells with specific inhibitors of the PI3K/Akt/mTOR pathway diminished T-cell apoptosis to levels seen after co-culture with wild-type PTEN tumor cells, suggesting that PTEN loss confers this immunoresistant phenotype through the PI3K/Akt/mTOR pathway. These results suggest that PTEN-deficient glioblastoma patients are suboptimal candidates for immunotherapy. In addition, our results raise the possibility of combining T-cell based immunotherapy protocols with clinical inhibitors of the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- James S Waldron
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California 94123, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
The detection of CMV pp65 and IE1 in glioblastoma multiforme. J Neurooncol 2010; 103:231-8. [PMID: 20820869 DOI: 10.1007/s11060-010-0383-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 08/21/2010] [Indexed: 12/17/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly lethal brain tumor affecting children and adults, with the majority of affected individuals dying from their disease by 2 years following diagnosis. Other groups have reported the association of cytomegalovirus (CMV) with GBM, and we sought to confirm these findings in a large series of patients with primary GBM from our institution. Immunohistochemical analysis of paraffin embedded tissue sections was performed on 49 newly diagnosed GBM tumors, the largest series reported to date. We confirmed the presence of CMV pp65 on 25/49 (51%) and of IE1 on 8/49 (16%) of these tumors. While pp65 and IE1 are generally found in the nucleus of cells that are permissibly infected by CMV, GBM in this series had mostly cytoplasmic staining, with only 16% having nuclear staining for one or both of these antigens. We infected GBM cell lines with a laboratory strain of CMV, and found that most of the staining was cytoplasmic, with some perinuclear localization of IE1. To test the potential for CMV infected GBM cells to be recognized by CMV pp65 and IE1 specific cytotoxic T lymphocytes (CTL), we used CMV infected GBM cell lines in cytotoxicity assays with human leukocyte antigen partially matched CMV CTL. Lysis of CMV infected GBM tumor cells was accentuated by pre-treating these cell lines with either the demethylating agent decitabine or interferon-γ, both of which were shown to increase MHC Class I and II expression on tumor cells in vitro. These studies confirm the presence of CMV pp65 or IE1 on approximately half of GBM, with the possibility that CMV positive tumor cells can be recognized by CMV pp65/IE1 specific T cells.
Collapse
|
44
|
CD8+ T-cell infiltrate in newly diagnosed glioblastoma is associated with long-term survival. J Clin Neurosci 2010; 17:1381-5. [PMID: 20727764 DOI: 10.1016/j.jocn.2010.03.031] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 03/23/2010] [Indexed: 11/20/2022]
Abstract
A growing body of evidence supports the significant interplay between the immune system and glioma pathogenesis. Here we investigate whether the extent of local glioma-associated CD8+ T-cell infiltrate at initial presentation correlates with long-term survival in patients with glioblastoma multiforme (GBM). The study was conducted by the University of California San Francisco Brain Tumor Research Center as part of the San Francisco Bay Area Adult Glioma Study, which included over 519 patients with GBM. A central neuropathology review was performed and populations of infiltrating CD8+ T-cells were quantified histologically. Of 108 patients studied, 43 patients had poor survival (<95days) and 65 patients had extended long-term survival of >403days. Tumors from long-term survivors were more likely than short-term survivors to have intermediate or extensive T-cell infiltrates compared to focal or rare infiltrates, and this association appears to be most significant in Caucasian women (p < 0.006). Thus, CD8+ T-cell infiltrate is associated with prolonged survival. Our data provide the impetus for more sophisticated studies to further elucidate prospectively the specific T-cell subtypes associated with long-term survival.
Collapse
|
45
|
Yang I, Han S, Parsa AT. Heat-shock protein vaccines as active immunotherapy against human gliomas. Expert Rev Anticancer Ther 2010; 9:1577-82. [PMID: 19895242 DOI: 10.1586/era.09.104] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Modern advances in cancer immunotherapy have led to the development of active immunotherapy that utilizes tumor-associated antigens to induce a specific immune response against the tumor. Current methods of immunotherapy implementation are based on the principle that tumor-associated antigens are capable of being processed by antigen-presenting cells and inducing an activated cytotoxic T-lymphocyte-specific immune response that targets the tumor cells. Antigen internalization and processing by antigen-presenting cells, such as dendritic cells, or macrophages results in their surface association with MHC class I molecules, which can be recognized by an antigen-specific cytotoxic T-lymphocyte adaptive immune response. With the aim of augmenting current immunotherapeutic modalities, much effort has been directed towards enhancing antigen-presenting cell activation and optimizing the processing of tumor-associated antigens and major histocompatibility molecules. The goal of these immunotherapy modifications is to ultimately improve the adaptive specific immune response in killing of tumor cells while sparing normal tissues. Immunotherapy has been actively studied and applied in glioblastomas. Preclinical animal models have shown the feasibility of an active immunotherapy approach through the utilization of tumor vaccines, and recently several clinical studies have also been initiated. Recently, endogenous heat-shock proteins have been implicated in the mediation of both the adaptive and innate immune responses. They are now being investigated as a potential modality and adjuvant to immunotherapy, and they represent a promising novel treatment for human glioblastomas.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
46
|
Gamma interferon-mediated superinduction of B7-H1 in PTEN-deficient glioblastoma: a paradoxical mechanism of immune evasion. Neuroreport 2010; 20:1597-602. [PMID: 19875977 DOI: 10.1097/wnr.0b013e32833188f7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
B7 homolog 1 (B7-H1) is a recently discovered immunoresistance protein that is regulated posttranscriptionally after PTEN loss in malignant glioma, a deadly form of brain tumor. Here, the impact of gamma-interferon-mediated activation of B7-H1 was investigated in glioblastoma patients with PTEN loss. Lymphocytes and T cells were selected for apoptosis assays after 1 : 1 coculture with autologous glioma cells. Gamma interferon treatment of PTEN-deficient tumors resulted in superinduction of B7-H1 protein that correlated with increased T-cell apoptosis, an effect dependent upon activation of the PI3-kinase pathway. The combination of PTEN loss and gamma-interferon exposure in glioblastoma patients results in an exceptionally immunoresistant phenotype that may negate adaptive immunity through induction of T-cell apoptosis.
Collapse
|
47
|
Han SJ, Kaur G, Yang I, Lim M. Biologic Principles of Immunotherapy for Malignant Gliomas. Neurosurg Clin N Am 2010; 21:1-16. [DOI: 10.1016/j.nec.2009.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
48
|
|
49
|
Abstract
Glioblastoma multiforme is the most common primary central nervous system tumor. The prognosis for these malignant brain tumors is poor, with a median survival of 14 months and a 5-year survival rate below 2%. Development of novel treatments is essential to improving survival and quality of life for these patients. Endogenous heat shock proteins have been implicated in mediation of both adaptive and innate immunity, and there is a rising interest in the use of this safe and multifaceted heat shock protein vaccine therapy as a promising treatment for human cancers, including glioblastoma multiforme.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurological Surgery, University of California at San Francisco, 505 Parnassus Avenue, Room M779, Campus 0112, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
50
|
Yang I, Huh NG, Smith ZA, Han SJ, Parsa AT. Distinguishing glioma recurrence from treatment effect after radiochemotherapy and immunotherapy. Neurosurg Clin N Am 2009; 21:181-6. [PMID: 19944976 DOI: 10.1016/j.nec.2009.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Recent advancements have made radiation and chemotherapy the standard of care for newly diagnosed glioblastomas. The use of these therapies has resulted in an increased diagnosis of pseudoprogression and radiation-induced necrosis. Standard MRI techniques are inadequate in differentiating tumor recurrence from posttreatment effects. Diagnosis of a posttreatment lesion as glioma recurrence rather than radiochemotherapy or immunotherapy treatment effect is critical. This increase in accuracy plays a role as newer immunotherapies incurring posttreatment effects on MRI emerge. Advancements with magnetic resonance spectroscopy, diffusion-weighted imaging, and functional positron emission tomography scans have shown promising capabilities. Further investigations are necessary to assess the imaging algorithms and accuracy of these modalities to differentiate true glioma recurrence from radiotherapy or immunotherapy treatment effect.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurological Surgery, University of California at San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA.
| | | | | | | | | |
Collapse
|