1
|
Luo N, Sun X, Ma S, Li X, Zhu W, Fu M, Yang F, Chen Z, Li Q, Zhang Y, Peng X, Hu G. Development of a Novel Prognostic Model of Glioblastoma Based on m6A-Associated Immune Genes and Identification of a New Biomarker. Front Oncol 2022; 12:868415. [PMID: 35936722 PMCID: PMC9348864 DOI: 10.3389/fonc.2022.868415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
Background Accumulating evidence shows that m6A regulates oncogene and tumor suppressor gene expression, thus playing a dual role in cancer. Likewise, there is a close relationship between the immune system and tumor development and progression. However, for glioblastoma, m6A-associated immunological markers remain to be identified. Methods We obtained gene expression, mutation, and clinical data on glioblastoma from The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. Next, we performed univariate COX–least absolute shrinkage and selection operator (LASSO)–multivariate COX regression analyses to establish a prognostic gene signature and develop a corresponding dynamic nomogram application. We then carried out a clustering analysis twice to categorize all samples according to their m6A-regulating and m6A-associated immune gene expression levels (high, medium, and low) and calculated their m6A score. Finally, we performed quantitative reverse transcription-polymerase chain reaction, cell counting kit-8, cell stemness detection, cell migration, and apoptosis detection in vitro assays to determine the biological role of CD81 in glioblastoma cells. Results Our glioblastoma risk score model had extremely high prediction efficacy, with the area under the receiver operating characteristic curve reaching 0.9. The web version of the dynamic nomogram application allows rapid and accurate calculation of patients’ survival odds. Survival curves and Sankey diagrams indicated that the high-m6A score group corresponded to the groups expressing medium and low m6A-regulating gene levels and high m6A-associated prognostic immune gene levels. Moreover, these groups displayed lower survival rates and higher immune infiltration. Based on the gene set enrichment analysis, the pathophysiological mechanism may be related to the activation of the immunosuppressive function and related signaling pathways. Moreover, the risk score model allowed us to perform immunotherapy benefit assessment. Finally, silencing CD81 in vitro significantly suppressed proliferation, stemness, and migration and facilitated apoptosis in glioblastoma cells. Conclusion We developed an accurate and efficient prognostic model. Furthermore, the correlation analysis of different stratification methods with tumor microenvironment provided a basis for further pathophysiological mechanism exploration. Finally, CD81 may serve as a diagnostic and prognostic biomarker in glioblastoma.
Collapse
Affiliation(s)
- Na Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xizi Sun
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengling Ma
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang, China
| | - Xiaoyu Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjun Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziqi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Guangyuan Hu, ; Xiaohong Peng, ; Yuanyuan Zhang,
| | - Xiaohong Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Guangyuan Hu, ; Xiaohong Peng, ; Yuanyuan Zhang,
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Guangyuan Hu, ; Xiaohong Peng, ; Yuanyuan Zhang,
| |
Collapse
|
2
|
Nduom EK, Weller M, Heimberger AB. Immunosuppressive mechanisms in glioblastoma. Neuro Oncol 2016; 17 Suppl 7:vii9-vii14. [PMID: 26516226 DOI: 10.1093/neuonc/nov151] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite maximal surgical and medical therapy, the treatment of glioblastoma remains a seriously vexing problem, with median survival well under 2 years and few long-term survivors. Targeted therapy has yet to produce significant advances in treatment of these lesions in spite of advanced molecular characterization of glioblastoma and glioblastoma cancer stem cells. Recently, immunotherapy has emerged as a promising mode for some of the hardest to treat tumors, including metastatic melanoma. Although immunotherapy has been evaluated in glioblastoma in the past with limited success, better understanding of the failures of these therapies could lead to more successful treatments in the future. Furthermore, there is a persistent challenge for the use of immune therapy to treat glioblastoma secondary to the existence of redundant mechanisms of tumor-mediated immune suppression. Here we will address these mechanisms of immunosuppression in glioblastoma and therapeutic approaches.
Collapse
Affiliation(s)
- Edjah K Nduom
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., A.B.H.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.)
| | - Michael Weller
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., A.B.H.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.)
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., A.B.H.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.)
| |
Collapse
|
3
|
Authier A, Farrand KJ, Broadley KW, Ancelet LR, Hunn MK, Stone S, McConnell MJ, Hermans IF. Enhanced immunosuppression by therapy-exposed glioblastoma multiforme tumor cells. Int J Cancer 2014; 136:2566-78. [DOI: 10.1002/ijc.29309] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 10/16/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Astrid Authier
- Vaccine Research Group; Malaghan Institute of Medical Research; Wellington 6242 New Zealand
| | - Kathryn J. Farrand
- Vaccine Research Group; Malaghan Institute of Medical Research; Wellington 6242 New Zealand
| | - Kate W.R. Broadley
- Vaccine Research Group; Malaghan Institute of Medical Research; Wellington 6242 New Zealand
| | - Lindsay R. Ancelet
- Vaccine Research Group; Malaghan Institute of Medical Research; Wellington 6242 New Zealand
| | - Martin K. Hunn
- Vaccine Research Group; Malaghan Institute of Medical Research; Wellington 6242 New Zealand
- School of Biological Sciences; Victoria University of Wellington; Wellington 6012 New Zealand
| | - Sarrabeth Stone
- School of Biological Sciences; Victoria University of Wellington; Wellington 6012 New Zealand
| | - Melanie J. McConnell
- School of Biological Sciences; Victoria University of Wellington; Wellington 6012 New Zealand
| | - Ian F. Hermans
- Vaccine Research Group; Malaghan Institute of Medical Research; Wellington 6242 New Zealand
- School of Biological Sciences; Victoria University of Wellington; Wellington 6012 New Zealand
| |
Collapse
|
4
|
Mechanisms of intimate and long-distance cross-talk between glioma and myeloid cells: how to break a vicious cycle. Biochim Biophys Acta Rev Cancer 2014; 1846:560-75. [PMID: 25453365 DOI: 10.1016/j.bbcan.2014.10.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/12/2014] [Accepted: 10/13/2014] [Indexed: 12/16/2022]
Abstract
Glioma-associated microglia and macrophages (GAMs) and myeloid-derived suppressor cells (MDSCs) condition the glioma microenvironment to generate an immunosuppressed niche for tumour expansion. This immunosuppressive microenvironment is considered to be shaped through a complex multi-step interactive process between glioma cells, GAMs and MDSCs. Glioma cells recruit GAMs and MDSCs to the tumour site and block their maturation. Glioma cell-derived factors subsequently skew these cells towards an immunosuppressive, tumour-promoting phenotype. Finally, GAMs and MDSCs enhance immune suppression in the glioma microenvironment and promote glioma growth, invasiveness, and neovascularization. The local and distant cross-talk between glioma cells and GAMs and MDSCs is regulated by a plethora of soluble proteins and cell surface-bound factors, and possibly via extracellular vesicles and platelets. Importantly, GAMs and MDSCs have been reported to impair the efficacy of glioma therapy, in particular immunotherapeutic approaches. Therefore, advancing our understanding of the function of GAMs and MDSCs in brain tumours and targeted intervention of their immunosuppressive function may benefit the treatment of glioma.
Collapse
|
5
|
Li W, Graeber MB. The molecular profile of microglia under the influence of glioma. Neuro Oncol 2012; 14:958-78. [PMID: 22573310 DOI: 10.1093/neuonc/nos116] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microglia, which contribute substantially to the tumor mass of glioblastoma, have been shown to play an important role in glioma growth and invasion. While a large number of experimental studies on functional attributes of microglia in glioma provide evidence for their tumor-supporting roles, there also exist hints in support of their anti-tumor properties. Microglial activities during glioma progression seem multifaceted. They have been attributed to the receptors expressed on the microglia surface, to glioma-derived molecules that have an effect on microglia, and to the molecules released by microglia in response to their environment under glioma control, which can have autocrine effects. In this paper, the microglia and glioma literature is reviewed. We provide a synopsis of the molecular profile of microglia under the influence of glioma in order to help establish a rational basis for their potential therapeutic use. The ability of microglia precursors to cross the blood-brain barrier makes them an attractive target for the development of novel cell-based treatments of malignant glioma.
Collapse
Affiliation(s)
- Wei Li
- Brain Tumor Research Laboratories, The Brain and Mind Research Institute, University of Sydney, 94 Mallett St, Camperdown, Sydney, NSW 2050, Australia
| | | |
Collapse
|
6
|
Kumar P, Acharya S, Chatterjee S, Kumari A, Chaudhuri S, Singh MK, Ghosh SN, Chaudhuri S. Immunomodulatory role of TIITS in respect to cytotoxic lymphocytes in four grades of human glioma. Cell Immunol 2012; 276:176-86. [DOI: 10.1016/j.cellimm.2012.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 05/11/2012] [Indexed: 11/16/2022]
|
7
|
Lorger M. Tumor microenvironment in the brain. Cancers (Basel) 2012; 4:218-43. [PMID: 24213237 PMCID: PMC3712675 DOI: 10.3390/cancers4010218] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 01/29/2012] [Accepted: 02/16/2012] [Indexed: 12/21/2022] Open
Abstract
In addition to malignant cancer cells, tumors contain a variety of different stromal cells that constitute the tumor microenvironment. Some of these cell types provide crucial support for tumor growth, while others have been suggested to actually inhibit tumor progression. The composition of tumor microenvironment varies depending on the tumor site. The brain in particular consists of numerous specialized cell types such as microglia, astrocytes, and brain endothelial cells. In addition to these brain-resident cells, primary and metastatic brain tumors have also been shown to be infiltrated by different populations of bone marrow-derived cells. The role of different cell types that constitute tumor microenvironment in the progression of brain malignancies is only poorly understood. Tumor microenvironment has been shown to be a promising therapeutic target and diagnostic marker in extracranial malignancies. A better understanding of tumor microenvironment in the brain would therefore be expected to contribute to the development of improved therapies for brain tumors that are urgently required due to a poor availability of treatments for these malignancies. This review summarizes some of the known interactions between brain tumors and different stromal cells, and also discusses potential therapeutic approaches within this context.
Collapse
Affiliation(s)
- Mihaela Lorger
- Leeds Institute of Molecular Medicine, University of Leeds, St. James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK.
| |
Collapse
|
8
|
Mechanisms of Immune Evasion by Gliomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:53-76. [DOI: 10.1007/978-1-4614-3146-6_5] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
9
|
Terasaka S, Kawabori M, Kobayashi H, Murata J, Kanno H, Tanaka S, Houkin K. Neurohypophyseal germinoma with abundant fibrous tissue. Brain Tumor Pathol 2011; 29:58-62. [PMID: 21918862 DOI: 10.1007/s10014-011-0063-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/13/2011] [Indexed: 12/14/2022]
Abstract
We report an unusual case of neurohypophyseal germinoma with abundant fibrous tissue and clival invasion that was initially misdiagnosed as lymphocytic hypophysitis. A 40-year-old woman presented with diabetes insipidus and panhypopituitarism after delivering her second son and which lasted for 4 years. Magnetic resonance imaging showed the intrasellar mass extending to the suprasellar region with enlarged pituitary stalk. The mass was heterogeneously enhanced and invaded the clivus. Biopsy of the intrasellar mass was performed via the trans-sphenoidal route, and histological examination revealed marked fibrous tissue and infiltration of lymphocytes, with no evidence of tumor cells. Lymphocytic hypophysitis was the initial diagnosis, and corticosteroid therapy was begun. Despite intensive treatment, the lesion enlarged and clinical symptoms worsened 2 weeks after surgery. Subtotal removal of the mass was performed, and a second histological examination revealed typical findings of the germinoma. Subsequently, the patient underwent chemoradiotherapy, and complete remission was achieved. Histological diagnosis is sometimes incorrect in fibrous tumors at the sellar region, and biopsy from several points is strongly recommended for this entity.
Collapse
Affiliation(s)
- Shunsuke Terasaka
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo, 060-8638, Japan
| | | | | | | | | | | | | |
Collapse
|
10
|
Overview of cellular immunotherapy for patients with glioblastoma. Clin Dev Immunol 2010; 2010. [PMID: 20953324 PMCID: PMC2952949 DOI: 10.1155/2010/689171] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/13/2010] [Accepted: 08/27/2010] [Indexed: 12/22/2022]
Abstract
High grade gliomas (HGG) including glioblastomas (GBM) are the most common and devastating primary brain tumours. Despite important progresses in GBM treatment that currently includes surgery combined to radio- and chemotherapy, GBM patients' prognosis remains very poor. Immunotherapy is one of the new promising therapeutic approaches that can specifically target tumour cells. Such an approach could also maintain long term antitumour responses without inducing neurologic defects. Since the past 25 years, adoptive and active immunotherapies using lymphokine-activated killer cells, cytotoxic T cells, tumour-infiltrating lymphocytes, autologous tumour cells, and dendritic cells have been tested in phase I/II clinical trials with HGG patients. This paper inventories these cellular immunotherapeutic strategies and discusses their efficacy, limits, and future perspectives for optimizing the treatment to achieve clinical benefits for GBM patients.
Collapse
|
11
|
Kushen MC, Sonabend AM, Lesniak MS. Current immunotherapeutic strategies for central nervous system tumors. Surg Oncol Clin N Am 2008; 16:987-1004, xii. [PMID: 18022555 DOI: 10.1016/j.soc.2007.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Immunotherapy has emerged as a promising tool in the management of malignant central nervous system tumors. Despite improvement in patient survival, traditional approaches, which consist mostly of surgery, radiotherapy, and chemotherapy, have been largely unsuccessful in permanently controlling these aggressive tumors. Immunotherapeutic strategies offer not only a novel approach but also an advantage in a way other modalities have been failing. Specifically, the capabilities of the immune system to recognize altered cells while leaving normal cells intact offer tremendous advantage over the conventional therapeutic approaches. This article summarizes our current understanding of immunotherapeutic treatment modalities used in clinical trials for management of malignant central nervous system tumors.
Collapse
Affiliation(s)
- Medina C Kushen
- Neurosurgical Oncology and The University of Chicago Brain Tumor Center, Section of Neurosurgery, The University of Chicago Hospital, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | | | | |
Collapse
|
12
|
Liu Y, Wang Q, Kleinschmidt-DeMasters BK, Franzusoff A, Ng KY, Lillehei KO. TGF-beta2 inhibition augments the effect of tumor vaccine and improves the survival of animals with pre-established brain tumors. J Neurooncol 2006; 81:149-62. [PMID: 16941073 DOI: 10.1007/s11060-006-9222-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Accepted: 07/17/2006] [Indexed: 10/24/2022]
Abstract
TGF-beta2 secretion by high grade gliomas has been implicated as one of the major factors contributing to tumor growth, alterations in the host immune response to tumor, and failure of gliomas to respond to current immunotherapy strategies. We hypothesized that targeted delivery and inhibition of TGF-beta2 by TGF-beta2 antisense oligonucleotides (AS-ODNs) would overcome tumor-induced immunosuppression and enhance the capacity of tumor vaccines to eradicate established brain tumors. Utilizing the mRNA sequences of TGF-beta2, specific AS-ODNs were constructed and tested for their ability to inhibit TGF-beta2 production in 9L glioma cells. The effect of combining local intracranial administration of antisense ODNs with systemic tumor vaccine was examined. Fisher 344 rats were vaccinated subcutaneously with irradiated 9L tumor cells 3 days after intracranial tumor implantation. Four days after vaccination, ODNs were administered into the tumor mass and survival was followed. ODNs delivered locally distributed widely within the brain tumor mass and inhibited TGF-beta2 expression. Survival of tumor-bearing rats treated with the combination of local antisense and systemic tumor vaccine was significantly enhanced (mean survival time (MST): 48.0 days). In contrast, MST for animals treated with nonsense plus vaccine, vaccine alone, antisense alone or PBS showed no survival advantage and no statistical differences between groups (33.5 days, 29.0 days, 37.5 days, and 31.5 days, respectively). Our data supports the hypothesis that local administration of antisense TGF-beta2 ODNs combined with systemic vaccination can increase efficacy of immunotherapy and is a novel, potentially clinically applicable, strategy for high-grade glioma treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurosurgery, C-307, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Denver, CO, 80262, USA
| | | | | | | | | | | |
Collapse
|
13
|
Hussain SF, Heimberger AB. Immunotherapy for human glioma: innovative approaches and recent results. Expert Rev Anticancer Ther 2006; 5:777-90. [PMID: 16221048 DOI: 10.1586/14737140.5.5.777] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The outcome for malignant glioma patients remains dismal despite treatment with surgical resection, radiation and chemotherapy. The goal of immunotherapy is to eradicate or suppress the residual infiltrative component of these tumors. Although there is clinical evidence for cell-mediated antiglioma activity, there are special considerations that need to be accounted for in the design of immunotherapeutics for CNS tumors, such as possible differences in antigen-presenting cells, trafficking of effector T-cells and immunosuppression. Previously characterized immunosuppression in glioma patients has included low peripheral blood lymphocyte counts, reduced delayed type hypersensitivity reactions to recall antigens, impaired mitogen-induced blastogenic responses by peripheral blood mononuclear cells, increased CD8+ suppressor T-cells, decreased CD4+ T-cell activity in vitro, diminished immunoglobulin synthesis by B-cells and impaired transmembrane signaling through the T-cell receptor/CD3 complex. Recent impairments that are being identified include anergy, failure of costimulation, lack of sufficient numbers of functional effector T-cells and the presence of T-suppressor cells within the tumor microenvironment. It is proposed that these inherent problems will need to be overcome in order for immunotherapies to realize their potential. Paradoxically, the efficacy of recent clinical immunotherapies for glioma patients appears equivalent to that seen in other cancer immunotherapeutic approaches. This review will provide an overview of the juxtaposition of the immune system and CNS, and will discuss the most recent and ongoing immunotherapeutic clinical trials that are demonstrating promising results.
Collapse
Affiliation(s)
- S Farzana Hussain
- University of Texas MD Anderson Cancer Center, Houston, TX 77230-1402, USA.
| | | |
Collapse
|
14
|
Sikorski CW, Lesniak MS. Immunotherapy for malignant glioma: current approaches and future directions. Neurol Res 2005; 27:703-16. [PMID: 16197807 DOI: 10.1179/016164105x49481] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Traditional therapies for the treatment of malignant glioma have failed to make appreciable gains regarding patient outcome in the last decade. Therefore, immunotherapeutic approaches have become increasingly popular in the treatment of this cancer. This article reviews general immunology of the central nervous system and the immunobiology of malignant glioma to provide a foundation for understanding the rationale behind current glioma immunotherapies. A review of currently implemented immunological treatments is then provided with special attention paid to the use of vaccines, gene therapy, cytokines, dendritic cells and viruses. Insights into future and developing avenues of glioma immunotherapy, such as novel delivery systems, are also discussed.
Collapse
Affiliation(s)
- Christian W Sikorski
- Division of Neurosurgery, The University of Chicago Pritzker School of Medicine, 5841 South Maryland Avenue, MC 3026, Chicago, Illinois 60637, USA
| | | |
Collapse
|
15
|
Abstract
Microglia play an important role in inflammatory diseases of the central nervous system (CNS). These cells have also been identified in brain neoplasms; however, as of yet their function largely remains unclear. More recent studies designed to characterize further tumor-associated microglia suggest that the immune effector function of these cells may be suppressed in CNS tumors. Furthermore, microglia and macrophages can secrete various cytokines and growth factors that may contribute to the successful immune evasion, growth, and invasion of brain neoplasms. A better understanding of microglia and macrophage function is essential for the development of immune-based treatment strategies against malignant brain tumors.
Collapse
Affiliation(s)
- Jyoti J Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53792-3232, USA
| | | | | |
Collapse
|
16
|
Zagzag D, Salnikow K, Chiriboga L, Yee H, Lan L, Ali MA, Garcia R, Demaria S, Newcomb EW. Downregulation of major histocompatibility complex antigens in invading glioma cells: stealth invasion of the brain. J Transl Med 2005; 85:328-41. [PMID: 15716863 DOI: 10.1038/labinvest.3700233] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Invasion into surrounding brain tissue is a fundamental feature of gliomas and the major reason for treatment failure. The process of brain invasion in gliomas is not well understood. Differences in gene expression and/or gene products between invading and noninvading glioma cells may identify potential targets for new therapies. To look for genes associated with glioma invasion, we first employed Affymetrix microarray Genechip technology to identify genes differentially expressed in migrating glioma cells in vitro and in invading glioma cells in vivo using laser capture microdissection. We observed upregulation of a variety of genes, previously reported to be linked to glioma cell migration and invasion. Remarkably, major histocompatiblity complex (MHC) class I and II genes were significantly downregulated in migrating cells in vitro and in invading cells in vivo. Decreased MHC expression was confirmed in migrating glioma cells in vitro using RT-PCR and in invading glioma cells in vivo by immunohistochemical staining of human and murine glioblastomas for beta2 microglobulin, a marker of MHC class I protein expression. To the best of our knowledge, this report is the first to describe the downregulation of MHC class I and II antigens in migrating and invading glioma cells, in vitro and in vivo, respectively. These results suggest that the very process of tumor invasion is associated with decreased expression of MHC antigens allowing glioma cells to invade the surrounding brain in a 'stealth'-like manner.
Collapse
Affiliation(s)
- David Zagzag
- Microvascular and Molecular Neuro-oncology Laboratory, New York University School of Medicine, New York, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Pinilla-Arias D, Mateo-Sierra O, Gutiérrez F, Fernández-Carballal C, Carrillo R. Inmunoterapia en astrocitomas de alto grado: principios y estado actual. Neurocirugia (Astur) 2005. [DOI: 10.1016/s1130-1473(05)70401-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Curtin JF, King GD, Candolfi M, Greeno RB, Kroeger KM, Lowenstein PR, Castro MG. Combining cytotoxic and immune-mediated gene therapy to treat brain tumors. Curr Top Med Chem 2005; 5:1151-70. [PMID: 16248789 PMCID: PMC1629031 DOI: 10.2174/156802605774370856] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is a type of intracranial brain tumor, for which there is no cure. In spite of advances in surgery, chemotherapy and radiotherapy, patients die within a year of diagnosis. Therefore, there is a critical need to develop novel therapeutic approaches for this disease. Gene therapy, which is the use of genes or other nucleic acids as drugs, is a powerful new treatment strategy which can be developed to treat GBM. Several treatment modalities are amenable for gene therapy implementation, e.g. conditional cytotoxic approaches, targeted delivery of toxins into the tumor mass, immune stimulatory strategies, and these will all be the focus of this review. Both conditional cytotoxicity and targeted toxin mediated tumor death, are aimed at eliminating an established tumor mass and preventing further growth. Tumors employ several defensive strategies that suppress and inhibit anti-tumor immune responses. A better understanding of the mechanisms involved in eliciting anti-tumor immune responses has identified promising targets for immunotherapy. Immunotherapy is designed to aid the immune system to recognize and destroy tumor cells in order to eliminate the tumor burden. Also, immune-therapeutic strategies have the added advantage that an activated immune system has the capability of recognizing tumor cells at distant sites from the primary tumor, therefore targeting metastasis distant from the primary tumor locale. Pre-clinical models and clinical trials have demonstrated that in spite of their location within the central nervous system (CNS), a tissue described as 'immune privileged', brain tumors can be effectively targeted by the activated immune system following various immunotherapeutic strategies. This review will highlight recent advances in brain tumor immunotherapy, with particular emphasis on advances made using gene therapy strategies, as well as reviewing other novel therapies that can be used in combination with immunotherapy. Another important aspect of implementing gene therapy in the clinical arena is to be able to image the targeting of the therapeutics to the tumors, treatment effectiveness and progression of disease. We have therefore reviewed the most exciting non-invasive, in vivo imaging techniques which can be used in combination with gene therapy to monitor therapeutic efficacy over time.
Collapse
Affiliation(s)
- James F Curtin
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Research Pavilion, Suite 5090, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Mukherjee J, Sarkar S, Ghosh A, Duttagupta AK, Chaudhuri S, Chaudhuri S. Immunotherapeutic effects of T11TS/S-LFA3 against nitrosocompound mediated neural genotoxicity. Toxicol Lett 2004; 150:239-57. [PMID: 15110076 DOI: 10.1016/j.toxlet.2004.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2003] [Revised: 01/05/2004] [Accepted: 01/20/2004] [Indexed: 10/26/2022]
Abstract
Nitrosocompounds formed by the interaction of nitrites and secondary amines are neurotoxic in human and different rodent species. Human exposure of nitrosocompounds are widespread affected by different modes like nitrite/nitrate preserved foods, beverages like beer, formed in the stomach following uptake of the precursors nitrates, nitrites and secondary amines. The productions of alkylating metabolites during the breakdown of nitrosocompounds are the causative agents for the neurotoxic changes of the neural cells. An attempt has been made in our lab to study the effect of nitrosocompound mediated toxicity and the gradual toxic effects of these neurotoxic agents to transform the normal glial cells to a neoplastic one. The present study indicated that a transmembrane glycopeptide of sheep red blood cell (SRBC), known as S-LFA3 or T11 target structure (T11TS) applied to nitrosocompound induced animals manifesting a full grown intracranial malignancy can revert back tumor-bearing condition to the normal physiological state. Young Druckray rat of both sexes aging 3-5 days were injected with N'-N'-ethyl nitrosourea (ENU) intraperitoneally (i.p.) at a single dose of 80 mg/kg body weight to simulate nitrosocompound mediated neurotoxicity. 2-,4-, 6-, 8-, and 10-month-old neonatal ENU induced animals were sacrificed for growth kinetics, functional immunological parameters and receptor studies to hint at the changes during tumor development. In order to determine the immunomodulatory role of T11TS, 7-month-old ENU induced animals were injected with T11TS at a dose of 0.41 mg/kg body weight, in three consecutive doses at an interval of 6 days maintaining normal control as untreated control and ENU induced animals of age-matched rats as tumor-bearing control. All the immunological parameters, growth kinetic study, receptor-based study by FACS directly established the immunomodulatory, anti-toxic and anti-tumor property of T11TS/S-LFA3. Finally, formation of DNA ladder along with the FACS-based apoptosis study clearly indicated that T11TS is a potent apoptotic inducer in neoplastic neural cells.
Collapse
Affiliation(s)
- Joydeep Mukherjee
- Department of Physiology, Cellular and Molecular Immunology Lab, Dr. B.C. Roy Post Graduate Institute of Basic Medical Sciences, Institute of Post Graduate Medical Education and Research, 244 B, J.C. Bose Road, Kolkata 700020, West Bengal, India
| | | | | | | | | | | |
Collapse
|
20
|
Kao JY, Gong Y, Chen CM, Zheng QD, Chen JJ. Tumor-derived TGF-beta reduces the efficacy of dendritic cell/tumor fusion vaccine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3806-11. [PMID: 12646647 DOI: 10.4049/jimmunol.170.7.3806] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dendritic cell (DC)-based antitumor vaccine is a novel cancer immunotherapy that is promising for reducing cancer-related mortality. However, results from early clinical trials were suboptimal. A possible explanation is that many tumors secrete immunosuppressive factors such as TGF-beta, which may hamper host immune response to DC vaccine. In this study, we demonstrated that TGF-beta produced by tumors significantly reduced the potency of DC/tumor fusion vaccines. TGF-beta-secreting (CT26-TGF-beta) stable mouse colon cancer cell lines were generated using a retroviral vector expressing TGF-beta. A non-TGF-beta-secreting (CT26-neo) cell line was generated using an empty retroviral vector. The efficacies of DC/tumor fusion vaccines were assessed in vitro and in vivo. DC/CT26-TGF-beta fusion cells failed to induce a strong T cell proliferative response in vitro, mainly due to the effect of TGF-beta on T cell responsiveness rather than DC stimulatory capability. Animals vaccinated with DC/CT26-TGF-beta fusion vaccine had lower tumor-specific CTL activity and had significantly lower survival after tumor challenge as compared with animals immunized with DC/CT26-neo hybrids (45 vs 77%, p < 0.05). Ex vivo exposure of DCs to TGF-beta did not appear to lessen the efficacy of DC vaccine. These data suggest that tumor-derived TGF-beta reduces the efficacy of DC/tumor fusion vaccine via an in vivo mechanism. Neutralization of TGF-beta produced by the fusion cells may enhance the effectiveness of DC-based immunotherapy.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Fractionation
- Cell-Free System/immunology
- Cell-Free System/metabolism
- Coculture Techniques
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Female
- Injections, Intraperitoneal
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred BALB C
- Neoplasm Proteins/adverse effects
- Neoplasm Proteins/blood
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/mortality
- Neoplasms, Experimental/prevention & control
- Suppressor Factors, Immunologic/adverse effects
- Suppressor Factors, Immunologic/blood
- Suppressor Factors, Immunologic/genetics
- Suppressor Factors, Immunologic/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transforming Growth Factor beta/administration & dosage
- Transforming Growth Factor beta/adverse effects
- Transforming Growth Factor beta/blood
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta1
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/transplantation
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- John Y Kao
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0650, USA
| | | | | | | | | |
Collapse
|
21
|
Yang L, Ng KY, Lillehei KO. Cell-mediated immunotherapy: a new approach to the treatment of malignant glioma. Cancer Control 2003; 10:138-47. [PMID: 12712008 DOI: 10.1177/107327480301000205] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The dismal prognosis for patients harboring intracranial gliomas has prompted an intensive search for effective treatment alternatives such as immunotherapy. Our increased knowledge in basic immunology, glioma immunobiology, and molecular biology may lead to the development of effective, rational immunotherapy approaches. METHODS The authors reviewed the literature on glioma immunology, the status of tumor vaccine therapy and on novel techniques to monitor the tumor-specific immune response. RESULTS Experimental conditions currently exist whereby potent antitumor cell-mediated immune responses can be generated. However, clinically, no therapeutic regimen has proven effective. Obstacles to establishing an effective immunotherapy regimen are the lack of a well-defined glioma-specific antigen, the heterogeneity of tumor cells in gliomas, and the modulating effect of the glioma itself on the immune system. Unique strategies to overcome these barriers are being developed. CONCLUSIONS Novel strategies to generate an anti-glioma immune response through use of dendritic cell vaccination, directed cytokine delivery, gene-based immunotherapy, and reversal of tumor-induced immunosuppression are promising. These strategies carry the potential of overcoming the resistance of gliomas to immunotherapeutic manipulation and, undoubtedly, will become a part of our future therapeutic armamentarium.
Collapse
Affiliation(s)
- Liu Yang
- Department of Neurosurgery, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | |
Collapse
|
22
|
Anderson RC, Elder JB, Brown MD, Mandigo CE, Parsa AT, Kim PD, Senatus P, Anderson DE, Bruce JN. Changes in the immunologic phenotype of human malignant glioma cells after passaging in vitro. Clin Immunol 2002; 102:84-95. [PMID: 11781071 DOI: 10.1006/clim.2001.5152] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although immunotherapeutic strategies against glioblastomas have been promising both in vitro and in animal models, similar successes have not been realized in human clinical trials. One reason may be that immunotherapeutic strategies are based on prior studies that primarily have used human glioblastoma cell lines passaged in vitro, which may not accurately reflect the in vivo properties of glioblastoma cells. In this report, we used flow cytometry to quantify the expression of immunological cell surface molecules on human glioblastomas directly ex vivo (prior to any in vitro culturing) and after varying passages in vitro. Furthermore, we used ELISA to quantitate cytokine secretion after various passages in vitro. We demonstrate that in vitro culturing of established cell lines led to increases in the cell surface expression of MHC class I and ICAM-1 and secretion of IL-6 and TGF-beta(2). Furthermore, there were significant changes in the expression of MHC class I, MHC class II, B7-2, ICAM-1, and FasL when comparing ex vivo tumor cells to those after a single passage in vitro. After passaging once in vitro, there were also significant changes in the secretion of TGF-beta(2) and IL-10. This report indicates that in vitro culturing leads to significant changes in both cell surface molecules and secreted cytokines, which are known to affect the ability of immune cells to initiate an anti-tumor immune response. These changes in the immunological phenotype of glioblastomas after in vitro culturing may in part explain the limited success of immunotherapeutic strategies against glioblastomas in human clinical trials.
Collapse
Affiliation(s)
- Richard C Anderson
- Department of Neurological Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Basse PH, Whiteside TL, Chambers W, Herberman RB. Therapeutic activity of NK cells against tumors. Int Rev Immunol 2001; 20:439-501. [PMID: 11878512 DOI: 10.3109/08830180109054416] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
While it is generally accepted that natural killer (NK) cells, by killing tumor cells in the circulation, represent a first line of defense against metastases, their therapeutic activity against established tumors has been limited. In this review, we describe studies to improve the therapeutic effectiveness of activated NK cells in both animal models and clinical trials to better understand the biological problems that limit their effectiveness.
Collapse
|
24
|
Liu Y, Ng K, Lillehei KO. Time course analysis and modulating effects of established brain tumor on active-specific immunotherapy. Neurosurg Focus 2000; 9:e3. [PMID: 16817686 DOI: 10.3171/foc.2000.9.6.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT There have been numerous attempts to establish an effective immunotherapy for the treatment of brain tumors. To date, reliable methods to manipulate the immune system for promoting brain tumor regression have been disappointing. Generation of active immune responses in most of these studies was only possible in the absence of viable tumor cells, suggesting that immunotherapy can only be used as preventive therapy. In few studies the investigators have demonstrated success in using immunotherapy to treat a preestablished intracranial tumor. Using the 9L intracranial glioma model, the authors sought to delineate the underlying mechanisms for these observations. METHODS In animals vaccinated with irradiated 9L glioma cells and interferon-gamma 14 and 7 days prior to intracranial tumor cell challenge, a significant increase in survival was shown. In contrast, vaccinations applied 3 days prior to, at the time of (Day 0) or 7 days after intracranial tumor cell challenge failed to influence survival. Histological examination of brain tissue specimens obtained in animals vaccinated before or after tumor cell challenge showed no difference in the degree of peritumoral mononuclear cell infiltration. When activated spleen cells obtained obtained from these animals were assayed for cytotoxicity and proliferative capacity, only those spleen cells derived from animals vaccinated prior to intracranial tumor cell challenge showed enhanced activity. CONCLUSIONS These data support the presence of a strong modulatory effect of tumor on local and systemic antitumoral immune response. This immunosuppression appears to be secondary to a direct effect on T-cell function. Reversal of this immunosuppression may be a useful adjunct to tumor vaccine therapy.
Collapse
Affiliation(s)
- Y Liu
- Department of Neurosurgery, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | |
Collapse
|
25
|
van Herpen CM, De Mulder PH. Locoregional immunotherapy in cancer patients: review of clinical studies. Ann Oncol 2000; 11:1229-39. [PMID: 11106110 DOI: 10.1023/a:1008335318764] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Many patients with invasive cancer have a compromised immune system. This immune dysfunction does appear to start at the site of the tumor. Locoregional immunotherapy is given to stimulate the immune system in order to kill tumor cells either indirectly via a specific or a non-specific way or directly via cell transfer therapy. Advantages to give this immunotherapy locoregionally in stead of systemically are a higher concentration of the immunomodulator at the site of the tumor, to attract or activate effector cells, and diminished toxicity. In this review we have summarised the clinical studies using loco-regional immunotherapy in patients with cancer. Only phase I and II studies were performed. Clinical responses were seen. No single locoregional treatment has become a standard therapy. Relatively few investigations were performed to estimate the influence on the locally effector mechanisms or immune dysfunction. In future clinical trials it is essential to get a better insight in these mechanisms.
Collapse
Affiliation(s)
- C M van Herpen
- Department of Medical Oncology, University Medical Centre Sint Radhoud, Nijmegen, The Netherlands.
| | | |
Collapse
|
26
|
Pollack IF, Okada H, Chambers WH. Exploitation of immune mechanisms in the treatment of central nervous system cancer. Semin Pediatr Neurol 2000; 7:131-43. [PMID: 10914414 DOI: 10.1053/pb.2000.6691] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Malignant gliomas are among the most common intrinsic brain tumors of both children and adults, and, because of unique aspects of their biology and anatomic site, they are the most refractory to conventional therapeutic strategies involving surgery, radiotherapy, or chemotherapy. Given the failure of standard therapies to improve the outlook of affected patients, significant attention has been focused on development of alternative treatments, particularly immunotherapy. Attempts have been made to treat gliomas using a variety of immunologically based strategies, including passive immunization, adoptive cellular immunotherapy, local and systemic delivery of biological response modifiers, and vaccination with tumor cells. Although preclinical modeling of these therapies provided an impetus for translation of their results into clinical protocols, these therapies have failed to yield consistently promising results in initial trials. However, significant insights into the immunobiology of the central nervous system (CNS) and gliomas have been gained from these studies, and have established that a number of immunobiological features of the brain and of gliomas themselves may be critical determinants in regulating efficacious treatment of these tumors. These include the following: (1) the presence of a blood-brain barrier that, although partially disrupted by the tumor, functions to exclude elements of the immune system from the tumor or brain parenchyma; (2) a lack of organized secondary lymphatic tissues supporting efficient immune responses locally in the CNS; (3) low levels of expression of major histocompatibility complex proteins in the CNS; (4) an apparent paucity of the most efficient antigen-presenting cells; and (5) glioma-derived immunosuppressive factors, such as transforming growth factor-beta, that interfere with the induction of local as well as systemic immune responses to the tumor. Recognition of these factors, and an appreciation of the underlying need for and validity of developing immunologically based therapies for gliomas, supports continued development of novel immunotherapeutic approaches, particularly those attempting to enhance the immunogenicity of glioma cells. This review addresses the current state of knowledge regarding the immunobiology of gliomas, recent developments in immunotherapy of gliomas, and promising future directions for development and implementation of cellular immunotherapy of gliomas.
Collapse
Affiliation(s)
- I F Pollack
- Department of Neurosurgery, University of Pittsburgh Cancer Institute Brain Tumor Center, Children's Hospital of Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
27
|
Hsieh CL, Chen DS, Hwang LH. Tumor-induced immunosuppression: a barrier to immunotherapy of large tumors by cytokine-secreting tumor vaccine. Hum Gene Ther 2000; 11:681-92. [PMID: 10757348 DOI: 10.1089/10430340050015581] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
An active immunotherapy strategy with cytokine-assisted tumor vaccine, although often effective for small tumor burdens, is much less so for large tumor burdens. This study examines how large tumors might suppress the T cell functions and escape from the immune responses elicited by a granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor vaccine. According to our results, the T cells isolated from the tumor-bearing mice treated late with the vaccine failed to confer protective activity on naive mice against a wild-type tumor challenge, unlike those isolated from the early-treated group. Nevertheless, the antitumor activity of the inactive T cells could be restored on in vitro stimulation. Expression of transforming growth factor beta (TGF-beta) and interleukin 10 (IL-10), the potent immunosuppressive factors, was detected in the parental tumor cell line RLmale 1 (a murine T leukemia cell line), as well as in the tumor region, the levels of which correlated with tumor progression. An in vitro assay of T cell functions revealed that the TGF-beta in the conditioned medium of RLmale 1 cells mainly affected the activation, whereas the IL-1male affected the activation to a lesser extent, but significantly affected the cytolytic activity, of tumor-specific T cells. The immunosuppressive activity of IL-10 was also signified by the findings that administration of the conditioned medium of RLmale 1 cultured in a serum-free medium, in which the TGF-beta activity was then lost while the IL-10 activity still remained, or of recombinant IL-10 to the early-treated group of mice abrogated the known efficacy of tumor vaccine on the small tumors. These data suggested that the efficacy of cytokine-secreting tumor vaccine was blocked by the immunosuppressive factors secreted from the large tumors. The results have important implications for the clinical design of immunotherapeutic strategies for advanced cancer patients.
Collapse
Affiliation(s)
- C L Hsieh
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei
| | | | | |
Collapse
|
28
|
Wagner S, Czub S, Greif M, Vince GH, Süss N, Kerkau S, Rieckmann P, Roggendorf W, Roosen K, Tonn JC. Microglial/macrophage expression of interleukin 10 in human glioblastomas. Int J Cancer 1999; 82:12-6. [PMID: 10360813 DOI: 10.1002/(sici)1097-0215(19990702)82:1<12::aid-ijc3>3.0.co;2-o] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Interleukin 10 (IL-10) expression has been found to be correlated with the extent of malignancy in gliomas. In vitro, IL-10 increases proliferation and migratory capacity in human glioma cell lines. In this study, we localized the site of IL-10 synthesis in gliomas to cells of microglial origin. Biopsy specimens from 11 patients with malignant glioma were processed on native tissues and at early cell culture passages (0-4). IL-10 mRNA was analyzed by RT-PCR and in situ hybridization. Protein was quantitatively assessed by ELISA in cell culture supernatants, and cells expressing IL-10 were determined by a combination of immunohistochemistry for CD68 (specific for microglia/macrophage lineage) and IL-10 in situ hybridization. IL-10 mRNA decreased from passage 0 to 4 in all samples and was undetectable beyond passage 5. Such downregulation of mRNA leads to a steep decrease of IL-10 protein in culture supernatants (below detection level, 0.05 ng/ml, beyond passage 1). The combination of in situ hybridization for IL-10 and CD68 immunostaining revealed that only cells of the microglia/macrophage lineage produced IL-10 mRNA. Our results identify microglia/macrophage cells as the major source of IL-10 expression in gliomas which decreases markedly during early passages of primary cultures of human gliomas due to a progressive reduction of microglia/macrophages present.
Collapse
Affiliation(s)
- S Wagner
- Department of Neurosurgery, University of Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
1. The identification of cytokine genes expressed in the central nervous system is critical to understanding the immune network in various diseases of brain, such as infection, degeneration, and malignancy. 2. Expression of cytokine genes in human astrocytoma cell lines and in fresh brain specimens was studied by the reverse-transcribed/polymerase chain reaction method. 3. The correlation between clinical malignancy and cytokine gene expression within malignant glioma was examined, especially regarding the relevancy of inhibitory cytokines, such as transforming growth factor-beta and interleukin-10.
Collapse
Affiliation(s)
- T Nitta
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Sasaki M, Plate KH. Gene therapy of malignant glioma: recent advances in experimental and clinical studies. Ann Oncol 1998; 9:1155-66. [PMID: 9862044 DOI: 10.1023/a:1008488709359] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent advances in molecular tumor biology and gene technology have provided the possibility to treat patients with malignant brain tumors by altering gene expression in tumor cells. Tumor development and progression involves alterations in a wide spectrum of genes, therefore a variety of gene therapy approaches for malignant gliomas have been proposed. In this review article, we discuss some principles of current gene therapeutic strategies that are under investigation in laboratories and in clinics. In addition, some general issues that remain to be resolved for clinical application of gene therapy in patients with malignant gliomas will be addressed.
Collapse
Affiliation(s)
- M Sasaki
- Department of Neuropathology, Freiburg University Medical School, Germany
| | | |
Collapse
|
31
|
Hsieh CL, Pang VF, Chen DS, Hwang LH. Regression of established mouse leukemia by GM-CSF-transduced tumor vaccine: implications for cytotoxic T lymphocyte responses and tumor burdens. Hum Gene Ther 1997; 8:1843-54. [PMID: 9382951 DOI: 10.1089/hum.1997.8.16-1843] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This study investigated the therapeutic effects of granulocyte-macrophage colony-stimulating factor (GM-CSF) on a mouse leukemia model. By using a retroviral vector, mouse GM-CSF cDNA was transduced into a highly tumorigenic T leukemia cell line, RL male 1. Injection of GM-CSF-secreting RL male 1 cells into syngeneic BALB/c mice elicited protective immunity in the animals, which could regress preestablished tumors introduced either by a subcutaneous or in an intravenous route. However, the therapeutic effects were less prominent in the mice inoculated with a large tumor load or in mice treated later. Winn tests further demonstrated that the splenocytes from the late-treated group conferred poorer protective effects in terms of reducing the growth of parental RL male 1 cells in naive mice than the splenocytes from the early-treated group. Nonetheless, upon stimulation in vitro, the activity of tumor-specific cytotoxic T lymphocytes (CTL) was comparable in the splenocytes of both groups of mice. Histological analysis also indicated that the CD8+ T cells appeared as early as 3 days following vaccination at the vaccine sites and at the tumor sites in both groups of mice. Above observations implied that the T cells in the animals bearing large tumors appeared to be in a state of suppression or anergy. Systematic histological analyses for 2 weeks provided further insight into various infiltrates at the vaccine sites and at the tumor sites in response to the inoculation of GM-CSF-secreting tumor vaccine.
Collapse
Affiliation(s)
- C L Hsieh
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei
| | | | | | | |
Collapse
|
32
|
Blömer U, Naldini L, Verma IM, Trono D, Gage FH. Applications of gene therapy to the CNS. Hum Mol Genet 1996; 5 Spec No:1397-404. [PMID: 8875243 DOI: 10.1093/hmg/5.supplement_1.1397] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Gene therapy is a new method with potential for treating a broad range of acquired and inherited neurologic diseases, where the causative gene defect or deletion has been identified. In addition to gene replacement the application of gene products that reduce cellular dysfunction or death represent new therapeutic options. Gene transfer techniques to express novel proteins using different viral vectors in vitro and in vivo, as well as animal models and human trials will be reviewed in this article. We will focus on a new lentiviral vector as a recent gene transfer method and degenerative disorders of the CNS, and their related model systems.
Collapse
Affiliation(s)
- U Blömer
- Salk Institute for Biological Studies, La Jolla, CA 92037-1099, USA
| | | | | | | | | |
Collapse
|
33
|
Hishii M, Nitta T, Ishida H, Ebato M, Kurosu A, Yagita H, Sato K, Okumura K. Human glioma-derived interleukin-10 inhibits antitumor immune responses in vitro. Neurosurgery 1995; 37:1160-6; discussion 1166-7. [PMID: 8584157 DOI: 10.1227/00006123-199512000-00016] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Intracranial malignant gliomas are sequestered from the immune system yet are associated with broad suppression of host immunocompetence. Immune system dysfunction in patients with gliomas seems to be related to inhibitory mediators produced by glioma cells. We investigated the physiological roles of glioma-derived interleukin (IL)-10 in Class II expression of monocytes, cytokine secretion from lymphocytes, and T cell proliferation in vitro. We could detect the messenger ribonucleic acid transcript of IL-10 in four gliomas by the reverse-transcribed polymerase chain reaction. Glioma-derived IL-10 greatly down-regulated human lymphocyte antigens-DR expression on monocytes. The inhibitory effect of IL-10 on interferon-gamma and tumor necrosis factor-alpha was neutralized by the anti-IL-10 monoclonal antibody; however, the inhibitory effect on IL-2 was not neutralized. Next, supernatants of glioma cells remarkably suppressed T cell proliferation in a dose-dependent fashion; however, this inhibitory effect was not restored by adding anti-IL-10 monoclonal antibodies. The supernatant also inhibited the allocytolytic activity of lymphocytes that were not neutralized by anti-IL-10 monoclonal antibody. IL-10 plays an important role in cytokine synthesis; nevertheless, impaired T cell responsiveness cannot be solely explained by glioma-derived IL-10.
Collapse
Affiliation(s)
- M Hishii
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Kaaijk P, Troost D, Dast PK, van den Berg F, Leenstra S, Bosch DA. Cytolytic effects of autologous lymphokine-activated killer cells on organotypic multicellular spheroids of gliomas in vitro. Neuropathol Appl Neurobiol 1995; 21:392-8. [PMID: 8632834 DOI: 10.1111/j.1365-2990.1995.tb01076.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Knowledge about lymphokine-activated killer (LAK) cell infiltration and LAK cell cytotoxicity is essential to improve the effectiveness of LAK cell therapy against gliomas. In the present study, organotypic multicellular spheroids (OMS) of glioma tissue were used as a culture model to study the effects of LAK cells on gliomas. Compared to tumour cell lines and spheroids derived from tumour cell lines, OMS have several advantages with respect to preservation of tumour cell heterogeneity and the maintenance of the tumour architecture, e.g. capillaries and extracellular matrix. Four glioma specimens, obtained at surgery, were cultured directly on agarose to form OMS, which were then co-cultured with either autologous LAK cells or autologous non-activated peripheral blood lymphocytes (PBLs). After various time periods of co-cultivation, the OMS were fixed and examined both histologically and immunocytochemically. The present results showed that LAK cells infiltrated the OMS completely within 24 h of co-cultivation and severe cellular damage was observed, whereas PBLs infiltrated the OMS poorly and there was only marginal cellular damage. The present study indicates that OMS of gliomas provide an experimental model to investigate the infiltration and cytotoxicity of LAK cells on glioma tissue in vitro.
Collapse
Affiliation(s)
- P Kaaijk
- Department of Neurosurgery, Academic Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
36
|
Weller M, Fontana A. The failure of current immunotherapy for malignant glioma. Tumor-derived TGF-beta, T-cell apoptosis, and the immune privilege of the brain. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1995; 21:128-51. [PMID: 8866671 DOI: 10.1016/0165-0173(95)00010-0] [Citation(s) in RCA: 172] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human malignant gliomas are rather resistant to all current therapeutic approaches including surgery, radiotherapy and chemotherapy as well as antibody-guided or cellular immunotherapy. The immunotherapy of malignant glioma has attracted interest because of the immunosuppressed state of malignant glioma patients which resides mainly in the T-cell compartment. This T-cell suppression has been attributed to the release by the glioma cells of immunosuppressive factors like transforming growth factor-beta (TGF-beta) and prostaglandins. TGF-beta has multiple effects in the immune system, most of which are inhibitory. TGF-beta appears to control downstream elements of various cellular activation cascades and regulates the expression of genes that are essential for cell cycle progression and mitosis. Since TGF-beta-mediated growth arrest of T-cell lines results in their apoptosis in vitro, glioma-derived TGF-beta may prevent immune-mediated glioma cell elimination by inducing apoptosis of tumor-infiltrating lymphocytes in vivo. T-cell apoptosis in the brain may be augmented by the absence of professional antigen-presenting cells and of appropriate costimulating signals. Numerous in vitro studies predict that tumor-derived TGF-beta will incapacitate in vitro-expanded and locally administered lymphokine-activated killer cells (LAK-cells) or tumor-infiltrating lymphocytes. Thus, TGF-beta may be partly responsible for the failure of current adoptive cellular immunotherapy of malignant glioma. Recent experimental in vivo studies on non-glial tumors have corroborated that neutralization of tumor-derived TGF-beta activity may facilitate immune-mediated tumor rejection. Current efforts to improve the efficacy of immunotherapy for malignant glioma include various strategies to enhance the immunogenicity of glioma cells and the cytotoxic activity of immune effector cells, e.g., by cytokine gene transfer. Future strategies of cellular immunotherapy for malignant glioma will have to focus on rendering glioma cell-targeting immune cells resistent to local inactivation and apoptosis which may be induced by TGF-beta and other immunosuppressive molecules at the site of neoplastic growth. Cytotoxic effectors targeting Fas/APO-1, the receptor protein for perforin-independent cytotoxic T-cell killing, might be promising, since Fas/APO-1 is expressed by glioma cells but not by untransformed brain cells, and since Fas/APO-1-mediated killing in vitro is not inhibited by TGF-beta.
Collapse
Affiliation(s)
- M Weller
- Neurologische Klinik der Universität Tübingen, Germany
| | | |
Collapse
|
37
|
Stulle K, Vollmers HP, Marquardt P, Müller-Hermelink HK. Human stomach carcinoma-specific T cells derived from the tumour-draining lymph nodes. Br J Cancer 1994; 70:1053-9. [PMID: 7981054 PMCID: PMC2033682 DOI: 10.1038/bjc.1994.448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In this paper we investigate the reactivity pattern of T cells from stomach carcinoma patients against autologous tumour cells. T cells obtained from the tumour environment, tumour-draining lymph nodes and peripheral blood were cloned in 78 patients with stomach cancer and anti-tumour cytotoxic T lymphocytes (CTLs) precursor frequencies were assessed in each sample by using limiting dilution analysis. When tumour-specific CTLs were tested for specific T-cell killing by using only low doses of Interleukin 2 (100 U ml-1), a moderate rate of proliferation frequency of T cells (0.047) and specific cytotoxicity (12%) were observed in lymph node populations. When both IL-2 and autologous tumour cells in mixed lymphocyte tumour cultures (MLTCs) were used for stimulation, a dramatic increase in number (0.1) and in specific lytic activity (46%) could be measured. No effect or specific activity to tumour cells was observed with peripheral blood lymphocytes and tumour-infiltrating lymphocytes.
Collapse
Affiliation(s)
- K Stulle
- Pathologisches Institut, Universität Würzburg, Germany
| | | | | | | |
Collapse
|
38
|
Jachimczak P, Schwulera U, Bogdahn U. In vitro studies of cytokine-mediated interactions between malignant glioma and autologous peripheral blood mononuclear cells. J Neurosurg 1994; 81:579-86. [PMID: 7931592 DOI: 10.3171/jns.1994.81.4.0579] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The humoral interactions between three malignant glioma early-passage cell cultures and in vitro interleukin (IL)-1 alpha- and IL-2-activated autologous peripheral blood mononuclear cells (PBMC's) were investigated, employing standard and modified (separated by permeable membranes) mixed lymphocyte tumor cell (MLTC) cultures. In modified MLTC's, glioma cells clearly inhibit proliferation of PBMC's (up to 60%), whereas lymphokine-activated PBMC's enhance glioma cell growth up to 12-fold, as determined by 3H-thymidine incorporation assays. Glioma cells produce both stimulatory (IL-6) and inhibitory proteins (transforming growth factor-beta) for PBMC's. Lymphokine-activated PBMC's secrete IL-1 alpha, IL-2, IL-4, IL-6, interferon-gamma, and tumor necrosis factor-alpha, which may modulate glioma cell proliferation. None of these cytokines stimulated glioma cells as intensely as modified MLTC systems. These observations indicate that in vitro lymphokine-activated PBMC's, although suppressed by humoral glioma-derived factors, may enhance glioma cell proliferation with soluble factors secreted into the culture medium. The authors conclude that glioma-lymphocyte growth regulatory networks include stimulatory and inhibitory factors from both cell populations, which may modulate tumor progression. These observations may have relevance for adoptive immunotherapy in patients with gliomas.
Collapse
Affiliation(s)
- P Jachimczak
- Department of Neurology, University of Würzburg, Germany
| | | | | |
Collapse
|
39
|
Boiardi A, Silvani A, Ruffini PA, Rivoltini L, Parmiani G, Broggi G, Salmaggi A. Loco-regional immunotherapy with recombinant interleukin-2 and adherent lymphokine-activated killer cells (A-LAK) in recurrent glioblastoma patients. Cancer Immunol Immunother 1994; 39:193-7. [PMID: 7923250 PMCID: PMC11038507 DOI: 10.1007/bf01533386] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/1994] [Accepted: 06/06/1994] [Indexed: 01/27/2023]
Abstract
Nine patients with recurrent glioblastoma were given autologous adherent lymphokine-activated killer (A-LAK) cells and interleukin-2 (IL-2) administered directly into the tumor cavity through an Ommaya tube placed during surgery/biopsy. The immunotherapy was well tolerated and the response rate was 33% (one complete response, two partial responses, four with stable disease and two with progressive disease). However, survival 18 months from initial diagnosis did not differ from that reported in the literature for patients treated conventionally. Serial determinations of IL-2 in the tumor cavity during the course of treatment revealed that IL-2 concentrations were sufficient to maintain lymphocyte activation. Since steroid medication was discontinued during treatment and A-LAK cells have greater antitumor activity than standard LAK cells, other factors are discussed that might explain the limited results.
Collapse
Affiliation(s)
- A Boiardi
- Istituto Nazionale Neurologico C. Besta, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
40
|
Nitta T, Hishii M, Sato K, Okumura K. Selective expression of interleukin-10 gene within glioblastoma multiforme. Brain Res 1994; 649:122-8. [PMID: 7953624 DOI: 10.1016/0006-8993(94)91055-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Little information exists regarding which glioma cells are able to escape immune system detection and progress within the host. In order to elucidate some of the mediators which facilitate the growth and spread of glioma cells, the expression of cytokines, TNF-alpha, IL-6, gamma-IFN, IL-10, and GM-CSF, within 12 human glioma specimens was investigated by the polymerase chain reaction. The twelve patients with malignant glioma were categorized into a localized (n = 4) and an invasive glioma (n = 8) groups, mostly glioblastoma multiforme, based upon the CT and MRI scans. We examined the correlation between specific cytokine gene expression and the clinical category of each patient. The results showed that while IL-10 mRNA transcripts were expressed in most of the tumors from the invasive glioma group (7/8), they were not expressed in tumors from the localized group. On the other hand, gamma-IFN gene expression was more frequent in tumors from the localized group (3/4 vs 1/8 from the invasive group). The mRNA transcripts of IL-6 and GM-CSF were more frequently expressed in tumors from the localized group. No consistent pattern was seen in TNF-alpha gene expression between the two groups. Among the five cytokines studied, IL-10 mRNA was selectively expressed within invasive gliomas compared to less malignant, localized glioma group. Our results demonstrate specific cytokine mRNA profiles in glioma patients, which might have prognostic significance for immunotherapy.
Collapse
Affiliation(s)
- T Nitta
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
41
|
Ram Z, Walbridge S, Heiss JD, Culver KW, Blaese RM, Oldfield EH. In vivo transfer of the human interleukin-2 gene: negative tumoricidal results in experimental brain tumors. J Neurosurg 1994; 80:535-40. [PMID: 8113867 DOI: 10.3171/jns.1994.80.3.0535] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The authors have recently shown the feasibility of eradicating brain tumors using in vivo retroviral-mediated transduction of tumors with the herpes simplex thymidine kinase (HStk) gene and ganciclovir therapy. However, thymidine kinase-transduced subcutaneous tumors in immunocompromised (athymic) mice were less responsive to this therapy than in immunocompetent animals, suggesting a role of the immune system in the process of tumor eradication. Broad suppression of humoral and cell-mediated immunity is found in patients with malignant gliomas. Interleukin-2 (IL-2) production and IL-2 receptor expression are decreased in gliomas patients. These findings and the proposed association between lymphocytic infiltration of brain tumors and survival suggest that immune response modifiers may be useful in treating glioma patients. To evaluate the role of local cytokine expression by tumor cells, alone or combined with HStk gene transfer and ganciclovir therapy, the authors investigated the efficacy of tumor (9L gliosarcoma) eradication in Fischer rats by in vitro and in vivo tumor transduction with the IL-2 gene alone or with a combined vector carrying both the HStk and IL-2 genes. Tumors injected with HStk vector-producer cells alone, with or without ganciclovir, and rats inoculated in the brain and subcutaneously with 9L cells that had previously been transduced in vitro served as controls. Murine vector-producer cells (3 x 10(6)/50 microliters) were injected into the brain tumors 7 days after tumor inoculation. Ganciclovir (15 mg/kg) was administered intraperitoneally twice daily for 10 days to animals that received HStk with or without IL-2 vector-producer cells, starting 5 days after producer-cell injection. The experiment was repeated with continuous daily treatment of all rats with oral dexamethasone (0.5 mg/kg). Rats were sacrificed 21 days after tumor inoculation, and the brains were removed for histological and immunohistochemical analysis for IL-2. Within each experimental group, tumors were found in a similar proportion in the dexamethasone-treated and untreated rats. Large brain tumors developed in all 10 rats that had been inoculated with 9L cells which had been pretransduced in vitro with the IL-2 gene, whereas only three of eight rats receiving subcutaneous inoculation of similar cells developed palpable tumors. No enhancement of tumor eradication was observed by adding the IL-2 gene in the HStk vector construct compared to the use of the vector with HStk alone. Lymphocytic infiltration was absent in all dexamethasone-treated rats but was observed in all treatment groups not receiving steroids.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- Z Ram
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | |
Collapse
|
42
|
Jachimczak P, Bogdahn U, Schneider J, Behl C, Meixensberger J, Apfel R, Dörries R, Schlingensiepen KH, Brysch W. The effect of transforming growth factor-beta 2-specific phosphorothioate-anti-sense oligodeoxynucleotides in reversing cellular immunosuppression in malignant glioma. J Neurosurg 1993; 78:944-51. [PMID: 8487077 DOI: 10.3171/jns.1993.78.6.0944] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This in vitro study was aimed at restitution of transforming growth factor (TGF)-beta 2-mediated suppression of T-lymphocyte activation within malignant gliomas. In early-passage tumor cell cultures of two glioblastomas (HTZ-153 and HTZ-209) and one malignant astrocytoma classified as World Health Organization Grade III (HTZ-243), autologous peripheral blood mononuclear cells were activated by interleukin-1 alpha and interleukin-2 in vitro (lymphokine-activated killer cells) and tested for cytotoxic and proliferative activity. In expression studies (Western blot and Northern hybridization) of all three tumors, TGF-beta could be detected at the protein and messenger ribonucleic acid (mRNA) levels. A polyclonal anti-TGF-beta neutralizing antibody did not enhance lymphocyte proliferation upon stimulation with tumor targets (3H-thymidine incorporation) and slightly stimulated lymphocyte cytotoxicity against autologous target cells. Preincubation of target cells for 12 hours with TGF-beta 2-specific phosphorothioate-anti-sense oligodeoxynucleotides (S-ODN's) did, however, enhance lymphocyte proliferation up to 2.5-fold and autologous tumor cytotoxicity up to 60%, compared to controls not treated with S-ODN's. Incubation of tumor cells with TGF-beta 2-specific S-ODN's resulted in decreased TGF-beta-specific immunoreactivity in cultured glioma cells, in reduced TGF-beta 2 protein concentration (Western blot), and in a change in the expression pattern of TGF-beta 2 mRNA's. These observations may have implications for in vivo and in vitro activation of a cellular immune response against autologous malignant glioma cells.
Collapse
MESH Headings
- Antigens, CD/analysis
- Antigens, Neoplasm/analysis
- Blotting, Northern
- Blotting, Western
- Brain Neoplasms/immunology
- Glioma/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunocompromised Host/drug effects
- Killer Cells, Lymphokine-Activated/immunology
- Leukocytes, Mononuclear/immunology
- Oligonucleotides, Antisense/pharmacology
- Phenotype
- RNA, Neoplasm/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- Transforming Growth Factor beta/drug effects
- Tumor Cells, Cultured/immunology
Collapse
Affiliation(s)
- P Jachimczak
- Department of Neurology, University of Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ruffini PA, Rivoltini L, Silvani A, Boiardi A, Parmiani G. Factors, including transforming growth factor beta, released in the glioblastoma residual cavity, impair activity of adherent lymphokine-activated killer cells. Cancer Immunol Immunother 1993; 36:409-16. [PMID: 8500113 PMCID: PMC11038209 DOI: 10.1007/bf01742258] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/1992] [Accepted: 12/15/1992] [Indexed: 01/31/2023]
Abstract
Adherent lymphokine-activated killer (A-LAK) cells were obtained from peripheral blood lymphocytes of patients with recurrent glioblastoma. In vitro features of A-LAK cultures were assessed in comparison to those of non-adherent lymphokine-activated killer (NA-LAK) cells of the same patients with regard to cytotoxic activity, proliferation and surface markers. Only in a minority of cases did A-LAK cells show a markedly higher cytotoxicity on K562, Daudi and allogeneic glioblastoma cells. Nevertheless, A-LAK cells proliferated significantly better than NA-LAK and contained higher percentages of CD16+, CD56+ and CD25+ cells, indicating that A-LAK cells from these patients represent a subpopulation of lymphocytes enriched for activated natural killer cells. We also investigated whether immunosuppressive factor(s) were present in the tumour bed of recurrent gliomas. To this end, samples of glioblastoma cavity fluid (GCF), which accumulates in the cavity of subtotally removed tumour, were recovered and tested for the presence of immunosuppressive activity. All GCF samples analysed were shown to inhibit in vitro proliferation and antitumour cytotoxicity of 1-week-cultured A-LAK cells in a dose-dependent manner. Such GCF activity was effectively antagonized by a transforming growth factor beta (TGF beta) neutralizing antibody, indicating the involvement of TGF beta in lymphocyte inhibition. These results show that in the tumour cavity remaining after subtotal glioblastoma resection a marked immunosuppressive activity, probably due to local release of TGF beta, is present; such activity may negatively influence the therapeutic effectiveness of local cellular immunotherapy.
Collapse
Affiliation(s)
- P A Ruffini
- Division of Experimental Oncology D, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | |
Collapse
|
44
|
Merlo A, Juretic A, Zuber M, Filgueira L, Lüscher U, Caetano V, Ulrich J, Gratzl O, Heberer M, Spagnoli GC. Cytokine gene expression in primary brain tumours, metastases and meningiomas suggests specific transcription patterns. Eur J Cancer 1993; 29A:2118-25. [PMID: 8297651 DOI: 10.1016/0959-8049(93)90046-i] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
To obtain an insight into the network of cytokine gene transcription in the brain tumour microenvironment, we investigated the expression of genes encoding for interleukin (IL)-1 alpha, IL-1 beta, IL-2, IL-4, IL-5, IL-6, IL-10, interferon (IFN)-gamma, granulocyte-macrophage colony-stimulating factor, tumour necrosis factor (TNF)-alpha and transforming growth factor (TGF)-beta 1, -beta 2 and -beta 3 in freshly excised brain tumour samples and autologous peripheral blood mononuclear cells. Tissue specimens from 15 primary brain tumours, three brain metastases, five meningiomas, autologous peripheral blood mononuclear cells (PBMC) and three brain tumour cell lines were tested by reverse polymerase chain reaction. Despite the presence of T-lymphocytes, cytokine gene transcripts typically detectable upon T cell receptor triggering could not be observed in central nervous system tumours of diverse histology. In primary brain neoplasms, transcription of genes encoding for the inhibitory cytokines TGF-beta and IL-10 was detectable in more than 50% of samples. IL-6 transcripts could only be detected in malignant gliomas. In brain metastases, virtually no cytokine gene transcripts could be observed. Surprisingly, TGF-beta transcripts were also detected in all meningiomas. Thus, transcription of genes encoding for inhibitory factors appears to prevail in primary brain neoplasms.
Collapse
Affiliation(s)
- A Merlo
- Department of Research, ZLF, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Li Y, Zhu C, Kong X, Zhang G. T subsets and antitumor activity of lymphocytes infiltrating human primary brain gliomas. Chin J Cancer Res 1992. [DOI: 10.1007/bf02996403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
46
|
Naganuma H, Sasaki A, Nukui H, Tasaka K. Antiproliferative cytokines secreted by lymphokine-activated killer cells stimulated with tumor cells. J Neurosurg 1992; 77:411-6. [PMID: 1506888 DOI: 10.3171/jns.1992.77.3.0411] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antiproliferative cytokine secretion by lymphokine-activated killer (LAK) cells during coculture with glioblastoma cell lines, autologous glioma cells, and nongliomatous tumor cell lines (Daudi and K562 cells) was assessed, as was the antiproliferative activity of the culture supernatants against the T98G (glioblastoma) cell line. A neutralization test using agents against interferon-gamma (IFN-gamma), tumor necrosis factor (TNF), and lymphotoxin (LT) showed that antiproliferative activity was due to IFN-gamma, but not to TNF or LT. Nongliomatous tumor cells stimulated LAK cells to secrete cytokines, but gliomatous tumor cells did not. It was found that there is a discrepancy between the LAK cell capability to lyse malignant glioma cells and the ability to secrete cytokines. This may be due to the factors secreted by glioblastoma cells.
Collapse
Affiliation(s)
- H Naganuma
- Department of Neurosurgery, Yamanashi Medical College, Japan
| | | | | | | |
Collapse
|
47
|
Myers RL, Whisler RL, Stephens RE, Sponseller CA, Livingston K, Spring PM, Yates AJ. Sensitivity of human glioma and brain cells to natural killer cell lysis. Effects of serum concentration, epidermal growth factor, and time in culture. J Neurosurg 1992; 76:986-90. [PMID: 1588434 DOI: 10.3171/jns.1992.76.6.0986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Using an in vitro monolayer natural killer (NK) cytolysis assay, the authors examined the effects of serum concentration and epidermal growth factor (EGF) on sensitivity to NK cytolysis. It was found that target cells cultured in high concentrations of serum (10% fetal bovine serum (FBS)) had higher cytotoxicity levels than those in low serum concentrations (0% to 0.5% FBS). Exposure of target cells to EGF had no effect on their sensitivity to NK cytolysis. Both glioma cell lines showed decreased NK cell sensitivity with longer times in culture. The results of cytofluorometric studies on these cell lines indicate that the differences in NK cell sensitivity may reflect the growth fraction of the target population and that a population with a higher proportion of cycling cells is more susceptible to lysis by NK cells. Whether it is possible to separate the proliferative rate of these cells from their NK cell sensitivity is unknown, but worthy of consideration.
Collapse
Affiliation(s)
- R L Myers
- Department of Pathology, Ohio State University, Columbus
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Patients with primary intracranial tumors (gliomas) exhibit a profound decrease in immunity, the mechanism of which has, until recently, remained obscure. Here Thomas Roszman, Lucinda Elliott and William Brooks reveal that T cells obtained from these patients exhibit defects in interleukin 2 secretion and in expression of the high-affinity IL-2 receptor and they discuss the role played by immunosuppressive factors produced by gliomas in inducing these defects.
Collapse
Affiliation(s)
- T Roszman
- Dept of Microbiology and Immunology, University of Kentucky Medical Center, Lexington 40536
| | | | | |
Collapse
|
49
|
Couldwell WT, Dore-Duffy P, Apuzzo ML, Antel JP. Malignant glioma modulation of immune function: relative contribution of different soluble factors. J Neuroimmunol 1991; 33:89-96. [PMID: 2066398 DOI: 10.1016/0165-5728(91)90052-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We analyzed a series of human glioma cell lines with regard to establishing what variables may contribute to their overall functional immunomodulating capability. We observed that supernatants derived from the gliomas, but not those from non-malignant human astrocyte cultures, suppressed lymphocyte proliferation. The extent of suppression elicited differed between tumors and for the same tumor depending upon its growth phase. For individual gliomas, supernatants from cultures approaching or at confluency elicited maximal lymphocyte suppression. For the series of tumors, levels of production of the immunosuppressive molecules transforming growth factor beta 2 and prostanoids (prostaglandin E2) did not correlate with the levels of functional suppression observed at any of the different growth phases. In some cases, glioma cultures grown in the presence of indomethacin to abolish prostanoid synthesis resulted in supernatants with net stimulatory activity. Our results indicate that malignant transformation of astrocytes is associated with acquisition of immunosuppressive capability which is determined by the combined effect of multiple immunomodulatory soluble factors, inhibitory or enhancing, and is dependent on the growth phase of the tumor.
Collapse
Affiliation(s)
- W T Couldwell
- Neuroimmunology Unit, McGill University, Montreal Neurological Institute and Hospital, Quebec, Canada
| | | | | | | |
Collapse
|