1
|
Dugandžija T, Drljača J, Bulajić D, Isaković A, Stilinović N, Sekulić S, Čapo I. Hallmarks of tumor-associated microglia response to experimental U87 human glioblastoma xenograft. Tissue Cell 2021; 72:101557. [PMID: 34051646 DOI: 10.1016/j.tice.2021.101557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is one of the deadliest primary brain neoplasm, heavily infiltrated with tumor-associated microglia/macrophages (TAM), which has received a great deal of interest. Bearing in mind that the number of peripheral macrophages by the 14th day is negligible, in our study TAM were referred to as microglia. Here we evaluated histopathological characterization of TAM and kinetics of their infiltration in U87 orthotopic GBM, a commonly used model in preclinical research. To mimic different stages of GBM growth, we evaluated three-time points. Our data showed that the highest areal density of TAM was 7 days after GBM inoculation, with ability to proliferate early after initiation of GBM growth. The areal density of TAM within the tumor correlated with GBM growth and proliferation. Moreover, microglia underwent substantial morphological changes upon exposure to GBM cells. A transition from ramified morphology in peritumoral area to ameboid shape with larger soma and shortened, thick branches in the tumor core was observed. Higher areal fraction of blood vessels also correlated with the areal density of TAM. Given these pro-invasive features of microglia, this GBM model represents a good basis for further testing microglia as a target and new strategy to fight with.
Collapse
Affiliation(s)
- Tihomir Dugandžija
- Faculty of Medicine, Department of Epidemiology, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia; Oncology Institute of Vojvodina, Put doktora Goldmana 4, Sremska Kamenica, 21204, Serbia
| | - Jovana Drljača
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia; Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia.
| | - Dragica Bulajić
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia; Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia
| | - Aleksandra Isaković
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Pasterova 2, Belgrade, 11000, Serbia
| | - Nebojša Stilinović
- Faculty of Medicine, Department of Pharmacology, Toxicology and Clinical Pharmacology, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia
| | - Slobodan Sekulić
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia; Department of Neurology, University Hospital, Clinical Center of Vojvodina, Hajduk Veljkova 1-7, Novi Sad, 21000, Serbia
| | - Ivan Čapo
- Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia; Faculty of Medicine, Department of Histology and Embryology, University of Novi Sad, Hajduk Veljkova 3, Novi Sad, 21000, Serbia
| |
Collapse
|
2
|
Hwang JS, Jung EH, Kwon MY, Han IO. Glioma-secreted soluble factors stimulate microglial activation: The role of interleukin-1β and tumor necrosis factor-α. J Neuroimmunol 2016; 298:165-71. [DOI: 10.1016/j.jneuroim.2016.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 12/13/2022]
|
3
|
Schijns VEJC, Pretto C, Devillers L, Pierre D, Hofman FM, Chen TC, Mespouille P, Hantos P, Glorieux P, Bota DA, Stathopoulos A. First clinical results of a personalized immunotherapeutic vaccine against recurrent, incompletely resected, treatment-resistant glioblastoma multiforme (GBM) tumors, based on combined allo- and auto-immune tumor reactivity. Vaccine 2015; 33:2690-6. [PMID: 25865468 PMCID: PMC10494870 DOI: 10.1016/j.vaccine.2015.03.095] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/10/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) patients have a poor prognosis. After tumor recurrence statistics suggest an imminent death within 1-4.5 months. Supportive preclinical data, from a rat model, provided the rational for a prototype clinical vaccine preparation, named Gliovac (or ERC 1671) composed of autologous antigens, derived from the patient's surgically removed tumor tissue, which is administered together with allogeneic antigens from glioma tissue resected from other GBM patients. We now report the first results of the Gliovac treatment for treatment-resistant GBM patients. Nine (9) recurrent GBM patients, after standard of care treatment, including surgery radio- and chemotherapy temozolomide, and for US patients, also bevacizumab (Avastin™), were treated under a compassionate use/hospital exemption protocol. Gliovac was given intradermally, together with human GM-CSF (Leukine(®)), and preceded by a regimen of regulatory T cell-depleting, low-dose cyclophosphamide. Gliovac administration in patients that have failed standard of care therapies showed minimal toxicity and enhanced overall survival (OS). Six-month (26 weeks) survival for the nine Gliovac patients was 100% versus 33% in control group. At week 40, the published overall survival was 10% if recurrent, reoperated patients were not treated. In the Gliovac treated group, the survival at 40 weeks was 77%. Our data suggest that Gliovac has low toxicity and a promising efficacy. A phase II trial has recently been initiated in recurrent, bevacizumab naïve GBM patients (NCT01903330).
Collapse
Affiliation(s)
- Virgil E J C Schijns
- Cell Biology & Immunology Group, Wageningen University, PO Box 338, 6700 AH Wageningen, The Netherlands; Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands.
| | - Chrystel Pretto
- Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands
| | - Laurent Devillers
- Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands
| | - Denis Pierre
- Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands
| | - Florence M Hofman
- Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | - Thomas C Chen
- Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA; Epitopoietic Research Corporation (ERC), 1055 E Colorado Blvd., Suite 500, Pasadena, CA 91106, USA
| | | | - Peter Hantos
- Department of Neurosurgery, Arlon and Libramont Hospital, Arlon and Libramont, Belgium
| | | | - Daniela A Bota
- Department of Neurology/Neurosurgery, University of California at Irvine, UC Irvine Medical Center, Irvine, CA, USA
| | - Apostolos Stathopoulos
- Cell Biology & Immunology Group, Wageningen University, PO Box 338, 6700 AH Wageningen, The Netherlands; Epitopoietic Research Corporation (ERC), Namur, Belgium; Epitopoietic Research Corporation (ERC), Schaijk, The Netherlands; Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA; Epitopoietic Research Corporation (ERC), 1055 E Colorado Blvd., Suite 500, Pasadena, CA 91106, USA; Department of Neurosurgery, Arlon and Libramont Hospital, Arlon and Libramont, Belgium; Department of Neurology/Neurosurgery, University of California at Irvine, UC Irvine Medical Center, Irvine, CA, USA.
| |
Collapse
|
4
|
Abstract
Despite dramatic advances in surgical techniques, imaging and adjuvant radiotherapy or chemotherapy, the prognosis for patients with malignant glial tumors remains dismal. Based on the current knowledge regarding immune responses in the healthy CNS and glioma-bearing hosts, this review discusses dendritic cell-based immunotherapeutic approaches for malignant gliomas and the relevance of recent clinical trials and their outcomes. It is now recognized that the CNS is not an immunologically tolerated site and clearance of arising glioma cells is a routine physiologic function of the normal, noncompromised immune system. To escape from immune surveillance, however, clinically apparent gliomas develop complex mechanisms that suppress tumoricidal immune responses. Although the use of dendritic cells for the treatment of glioma patients may be the most appropriate approach, an effective treatment paradigm for these tumors may eventually require the use of several types of treatment. Additionally, given the heterogeneity of this disease process and an immune-refractory tumor cell population, the series use of rational multiple modalities that target disparate tumor characteristics may be the most effective therapeutic strategy to treat malignant gliomas.
Collapse
Affiliation(s)
- Yasuharu Akasaki
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Suite 800 East, 8631 West 3 Street, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
5
|
Abstract
There is increasing evidence in the literature pointing to an important role of inflammation during initiation and progression of cancer. Glioblastoma is the most common malignant primary brain tumor with approximately 23,000 newly-diagnosed cases each year in the United States, and has a dismal median survival of only 15 months. Although the blood-brain barrier maintains an immune-privileged status of the brain under steady state, intracranial tumors including gliomas are invariably infiltrated with various types of immune cells. The T helper 17 (Th17) cells, a recently discovered interleukin (IL)-17-producing T cell subtype, have been reported in several extracranial and some intracranial tumors, where they have been implicated in either pro- or antitumor activity depending on the tumor type. Here, we present a succinct review of the current literature on the prevalence and potential role of IL-17 in malignant gliomas. Further mechanistic studies on IL-17 mediated inflammatory pathway in gliomas may provide with opportunities for novel immunotherapeutic interventions.
Collapse
Affiliation(s)
- Prahlad Parajuli
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University and Karmanos Cancer Institute, Detroit, USA
| |
Collapse
|
6
|
Tada M, de Tribolet N. Immunobiology of malignant gliomas. J Clin Neurosci 2012; 3:102-13. [PMID: 18638850 DOI: 10.1016/s0967-5868(96)90001-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/1995] [Accepted: 05/25/1995] [Indexed: 12/19/2022]
Abstract
The immune system of patients with malignant gliomas is profoundly suppressed. The suppression involves both the cellular and humoral immunity and it is mainly attributable to selective depletion and malfunction of helper T cells. Malignant glioma cells express potent immunosuppressive factors such as transforming growth factor-beta(2), inteleukin-10 and prostaglandin E(2). Malignant glioma cells also produce chemoattractants and immunostimulatory cytokines which may activate the immune cells. However, the production of these stimulatory cytokines is not self-destructive to glioma cells because of the immunosuppression. Rather, the tumour cells use them to gain a growth advantage. Indeed the cytokines may act as a growth stimulator of the tumour cells themselves (autocrine mechanism), they may act as angiogenic factors to endothelial cells (paracrine mechanism) or induce the attracted immune cells to secrete angiogenic factors. Some cytokines produced by malignant glioma cells are known to be growth inhibitory to normal astrocytes. Recent studies on tumour suppressor genes suggest a close link between the aberrant genes and the immunobiologic features of malignant glioma cells.
Collapse
Affiliation(s)
- M Tada
- Department of Neurosurgery, University Hospital, Lausanne, Switzerland
| | | |
Collapse
|
7
|
Paladugu M, Thakur A, Lum LG, Mittal S, Parajuli P. Generation and immunologic functions of Th17 cells in malignant gliomas. Cancer Immunol Immunother 2012; 62:75-86. [PMID: 22752645 DOI: 10.1007/s00262-012-1312-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/15/2012] [Indexed: 12/22/2022]
Abstract
Th17 cells, a recently discovered inflammatory T cell subtype, have been implicated with autoimmune disorders. However, mechanism of generation or functions of intratumoral Th17 cells are still unclear. We have been investigating the mechanism of induction and role of Th17 cells in malignant gliomas using primary tumor as well as cell lines. We report here that: (1) a higher frequency of Th17 cells in gliomas were associated with higher number of myeloid (CD11b) cells as well as the expression of TGF-β1 or IL-6; (2) conditioned medium from glioma cells (Gl CM) induced Th17 cell differentiation, which was inhibited by anti-TGF-β1 and anti-IL-6; (3) glioma-associated monocytes secreted Th17-promoting cytokines IL-1β and IL-23; (4) CM from glioma and monocyte co-culture (Gl+Mo CM) induced high frequency of Th17 cells in naïve T cell culture, which was abrogated by anti-IL-1β and anti-IL-23 antibodies; (5) In vitro Gl+Mo CM-mediated Th17 generation was associated with a decrease in IFN-γ and a concomitant increase in IL-10 secretion. Anti-TGF-β1, but not anti-IL-6, significantly reversed this cytokine profile. These results demonstrate prevalence of Th17 cells in gliomas and implicate the cytokines derived from the tumor as well as infiltrating myeloid cells in the induction of Th17 cells in glioma microenvironment. Moreover, the data also suggest that glioma-associated Th17 cells may contribute to immune-suppression via TGF-β1-induced IL-10 secretion. Further studies on the mechanism of tumor-infiltration, developmental pathways, and pro-/anti-tumor functions of Th17 cells will provide rationale for developing novel adjuvant immunotherapeutic strategies for malignant gliomas.
Collapse
Affiliation(s)
- Manjeera Paladugu
- Department of Neurosurgery, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
8
|
Beauchesne P. Extra-neural metastases of malignant gliomas: myth or reality? Cancers (Basel) 2011; 3:461-77. [PMID: 24212625 PMCID: PMC3756372 DOI: 10.3390/cancers3010461] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 01/04/2011] [Accepted: 01/19/2011] [Indexed: 01/15/2023] Open
Abstract
Malignant gliomas account for approximately 60% of all primary brain tumors in adults. Prognosis for these patients has not significantly changed in recent years-despite debulking surgery, radiotherapy and cytotoxic chemotherapy-with a median survival of 9-12 months. Virtually no patients are cured of their illness. Malignant gliomas are usually locally invasive tumors, though extra-neural metastases can sometimes occur late in the course of the disease (median of two years). They generally appear after craniotomy although spontaneous metastases have also been reported. The incidence of these metastases from primary intra-cranial malignant gliomas is low; it is estimated at less than 2% of all cases. Extra-neural metastases from gliomas frequently occur late in the course of the disease (median of two years), and generally appear after craniotomy, but spontaneous metastases have also been reported. Malignant glioma metastases usually involve the regional lymph nodes, lungs and pleural cavity, and occasionally the bone and liver. In this review, we present three cases of extra-neural metastasis of malignant gliomas from our department, summarize the main reported cases in literature, and try to understand the mechanisms underlying these systemic metastases.
Collapse
Affiliation(s)
- Patrick Beauchesne
- Neuro-Oncology, CHU de NANCY, Hôpital Central, CO n°34, 54035 Nancy Cedex, France.
| |
Collapse
|
9
|
Samaras V, Piperi C, Levidou G, Zisakis A, Kavantzas N, Themistocleous MS, Boviatsis EI, Barbatis C, Lea RW, Kalofoutis A, Korkolopoulou P. Analysis of interleukin (IL)-8 expression in human astrocytomas: associations with IL-6, cyclooxygenase-2, vascular endothelial growth factor, and microvessel morphometry. Hum Immunol 2009; 70:391-7. [PMID: 19332096 DOI: 10.1016/j.humimm.2009.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 03/11/2009] [Accepted: 03/24/2009] [Indexed: 01/09/2023]
Abstract
Malignant astrocytomas are highly vascular neoplasms with potent angiogenic activity. The present study aimed to investigate peripheral and local expression of interleukin (IL)-8 in astrocytomas with possible associations to IL-6, cyclooxygenase-2 (COX-2), vascular endothelial growth factor (VEGF) expression, and microvessel morphometry. IL-6- and IL-8-secreting peripheral blood monocytes (PBMCs) were evaluated in 17 glioblastoma (WHO grade IV), 5 anaplastic astrocytoma (WHO grade III), and 6 diffuse astrocytoma patients (WHO grade II), in parallel with 23 healthy controls using enzyme-linked immunosorbent spot (ELISPOT) assay. The IL-8 expression was assessed immunohistochemically in patients' tumor tissue sections and correlated with the expression of COX-2, VEGF, IL-6, and microvessel morphometry (assessed using CD34 antibody). Eighteen cases were also stained for CD31 and used as an additional vessel marker to validate our results regarding microvessel morphometry. IL-6 and IL-8 were highly secreted in the PBMCs of glioma patients compared with controls (p = 0.0001, p < 0.0001, respectively), with a positive correlation between IL-8 expression and secretion levels (p = 0.001). IL-8 immunoreactivity was detected in malignant cells or macrophages in perivascular areas and in pseudopalisading cells around necrosis and was positively correlated with histological grade (p = 0.0175) and tumor necrosis (p = 0.0793). IL-6 and IL-8 expression levels were positively correlated (p = 0.0036) and associated with COX-2 and VEGF expression (IL-6: p = 0.0133, p = 0.065; IL-8: p = 0.0139, p = 0.0101), but not with microvessel morphometry, by either CD31 or CD34. The coordinate expression and topographical relationship of IL-6, IL-8, COX-2, and VEGF in the same tumor areas (e.g., perinecrotic areas) attest to their intimate liaison in terms of cancer-induced angiogenesis, which is probably secondary to the induction of multiple interdependent molecular pathways. Moreover, our study seems to be the first attempt to link IL-8 expression by tumor cells with histological grade, implicating its potent role in gliomagenesis.
Collapse
Affiliation(s)
- Vassilis Samaras
- Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Laiko Hospital, Athens 11527, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Tanriover N, Ulu MO, Sanus GZ, Bilir A, Canbeyli R, Oz B, Akar Z, Kuday C. The effects of systemic and intratumoral interleukin-12 treatment in C6 rat glioma model. Neurol Res 2009; 30:511-7. [PMID: 18953742 DOI: 10.1179/174313208x289516] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Cytokine based immunotherapy has long been an exciting field for many investigators aiming to provide an effective alternative treatment modality for glioma management. Among these cytokines, interleukin-12 (IL-72) plays a crucial role in mediating inflammatory and antitumoral activity on the host defence. We have investigated the therapeutic role of systemic and local delivery of IL-12 in C6 rat glioma model and compared these two modalities. METHODS The donor C6 glioma cells were injected stereotactically to 32 Wistar rats and right frontal tumor formation was established in all subjects. The rats were evenly divided into four groups as intratumoral (i.t.) control group (Group IA), intraperitoneal (i.p.) control group (Group IB), i.t. treatment group (Group II) and i.p. treatment group (Group III). Magnetic resonance imaging were performed to 72 rats (three from each group) on the seventh post-inoculation day. Recombinant mouse IL-12 (rmIL-12) was administered via i.t. (0.1 microg 5 microl/day/rat) and i.p. (0.1 microg 20 microl/day/rat) routes to treatment groups between days 9 and 11 following tumor inoculation, for 3 consecutive days. The rats which were unresponsive to the external stimuli, unable to feed themselves or having severe neurological impairment were decapitated and the specimens were histopathologically examined. RESULTS The subjects of Group ILL (i.p.) showed a statistically significant prolongation in survival time (mean = 39 days) when compared to the control group (mean = 31.7 days) (p = 0.035) and Group II (i.t.) (mean = 24.5 days) (p = 0.005). Histopathologic examination of Group III revealed markedly increased intratumoral and peritumoral lymphocyte infiltration compared with the other groups. CONCLUSION This study demonstrated that systemic administration of IL- 12 in C6 glioma model in rats prolongs the survival, probably by stimulating the cellular immunity leading to lymphocytic infiltration.
Collapse
Affiliation(s)
- Necmettin Tanriover
- Department of Neurosurgery and Department of Pathology, Cerrahpasa Medical Faculty, Istanbul.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Mariani CL, Rajon D, Bova FJ, Streit WJ. Nonspecific immunotherapy with intratumoral lipopolysaccharide and zymosan A but not GM-CSF leads to an effective anti-tumor response in subcutaneous RG-2 gliomas. J Neurooncol 2007; 85:231-40. [PMID: 17568998 DOI: 10.1007/s11060-007-9415-2] [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: 03/06/2007] [Accepted: 05/11/2007] [Indexed: 01/09/2023]
Abstract
PURPOSE Nonspecific stimulation of cells of the immune system may be useful in generating an anti-tumor response for a variety of cancers and may work synergistically with currently available cytotoxic therapies. In this study we examined the response of syngeneic rat gliomas to treatment with several nonspecific stimulators of dendritic cells and macrophages alone or in combination with radiation therapy. EXPERIMENTAL DESIGN RG-2 gliomas were implanted subcutaneously and treated with intratumoral (IT) injections of the toll-like receptor (TLR) ligands lipopolysaccharide (LPS) and zymosan A (ZymA) and the cytokine granulocyte-macrophage colony stimulating factor (GM-CSF). Combination treatment with IT LPS and single-fraction external beam radiotherapy (EBRT) was also evaluated. RESULTS Treatment with IT LPS and ZymA delayed tumor growth compared to saline controls. Multiple doses of both substances were superior to single doses, and led to complete tumor regression in 71% (LPS) and 50% (ZymA) of animals. GM-CSF showed no anti-tumor effects in this study. Combinations of IT LPS and EBRT appeared to have a synergistic effect in delaying tumor growth. Rechallenge studies and IT LPS treatment of RG-2 tumors in nude rats suggested the importance of T cells in this treatment paradigm. CONCLUSIONS Direct IT treatment with the TLR ligands LPS and ZymA are effective in generating an anti-tumor response. These treatments may synergize with cytotoxic therapies such as EBRT, and appear to require T cells for a successful outcome.
Collapse
Affiliation(s)
- Christopher L Mariani
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA.
| | | | | | | |
Collapse
|
12
|
Samaras V, Piperi C, Korkolopoulou P, Zisakis A, Levidou G, Themistocleous MS, Boviatsis EI, Sakas DE, Lea RW, Kalofoutis A, Patsouris E. Application of the ELISPOT method for comparative analysis of interleukin (IL)-6 and IL-10 secretion in peripheral blood of patients with astroglial tumors. Mol Cell Biochem 2007; 304:343-51. [PMID: 17551671 DOI: 10.1007/s11010-007-9517-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Accepted: 05/16/2007] [Indexed: 02/07/2023]
Abstract
Glioblastoma, (grade IV astrocytoma), is characterized by rapid growth and resistance to treatment. Identification of markers of aggressiveness in this tumor could represent new therapeutic targets. Interleukins (IL)-6 and IL-10 may be considered as possible candidates, regulating cell growth, resistance to chemotherapy and angiogenesis. ELISPOT method provides a useful tool for the determination of the exact cell number of peripheral lymphocytes secreting a specific cytokine. IL-6 and IL-10 secretion levels were determined using ELISPOT methodology in peripheral blood mononuclear cells of 18 patients with astrocytic neoplasms (3 grade II and 15 grade IV), in parallel with 18 healthy controls. Additionally, immunohistochemical expression of these two cytokines was performed in paraffin-embedded neoplastic tissue in 12 of these patients. The secretion of IL-6 from peripheral monocytes was significantly higher in glioma patients compared to controls (P = 0.0003). In addition, IL-10 secretion from peripheral mononuclear and tumor cells of glioma patients was also higher as compared to healthy controls (P = 0.0002). Based on immunohistochemical staining, IL-6 expression was localized in tumor cells and macrophages as well as in areas of large ischemic necrosis, while the major source of IL-10 expression in glioblastomas was the microglia/macrophage cells. It is suggested that IL-10 contributes to the progression of astrocytomas by suppressing the patient's immune response, whereas IL-6 provides an additional growth advantage. This study demonstrates for the first time the usefulness of ELISPOT in estimating the secretion of IL-6 and IL-10 from peripheral blood and the correlation of their expression in neoplastic cells.
Collapse
Affiliation(s)
- Vassilis Samaras
- Department of Pathology, University of Athens Medical School, M.Asias 75, Goudi 11527, Athens, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Parajuli P, Mathupala S, Mittal S, Sloan AE. Dendritic cell-based active specific immunotherapy for malignant glioma. Expert Opin Biol Ther 2007; 7:439-48. [PMID: 17373896 DOI: 10.1517/14712598.7.4.439] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Immunotherapy is an appealing therapeutic modality for malignant gliomas because of its potential to selectively target residual tumor cells that have invaded the normal brain. Most immunotherapeutic studies are designed to exploit the capacity of dendritic cells for inducing cell-mediated effects as well as immune memory responses for destroying residual tumor cells and preventing recurrence. Although initial clinical studies on dendritic cell-based immunotherapy resulted in very limited success, they have prompted many new studies on exploring strategies to induce a more robust antitumor immune response by using novel adjuvants for maturation and activation of dendritic cells. More studies have focused on the mechanisms of immune suppression by tumor cells and the role of regulatory T cells in tumor growth and progression. In this article, the authors review the evolution of dendritic cell-based immunotherapeutic strategies for adjuvant treatment of malignant gliomas. The authors also discuss how new knowledge on tumor-intrinsic mechanisms of tolerance induction and immunosuppression are likely to shape the future of immunotherapy for high-grade gliomas.
Collapse
Affiliation(s)
- Prahlad Parajuli
- Wayne State University and Karmanos Cancer Institute, Department of Neurosurgery, Hudson-Webber CRC #808, 4100 John R St, Detroit, MI-48201, USA.
| | | | | | | |
Collapse
|
14
|
Nasser JA, Falavigna A, Ferraz F, Duigou G, Bruce J. Transcription analysis of TIMP-1 and NM23-h1 genes in glioma cell invasion. ARQUIVOS DE NEURO-PSIQUIATRIA 2006; 64:774-80. [PMID: 17057884 DOI: 10.1590/s0004-282x2006000500014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 05/25/2006] [Indexed: 11/21/2022]
Abstract
PURPOSE: To evaluate using transcription analysis the presence and importance of two genes: NM23-H1 and TIMP-1 on control of tumor cell invasion in diffuse astrocytomas (WHO II) and glioblastoma multiforme (WHO IV). METHOD: Northern blot analysis of NM23-H1 and TIMP-1 was performed. Eight diffuse astrocytomas and 19 glioblastomas (WHO IV) were analyzed to determine if TIMP-1 and NM23-H1 were candidates to inhibition of tumor cell invasion quantitated RNA levels. The samples were collected directly from operating room. Total cellular RNA was extracted from frozen tissue samples using guanidinium-isothiocyanate and cesium chloride gradients. Total RNA (10 mg per sample) from tumor tissue were size fractionated through 1% agarose-formaldehyde gel and transferred to nylon filters and then hybridized to 32P-labeled DNA probes and placed for autoradiography. Levels of specific RNAs were determined by computer-assisted laser densitometry. Blot filters were sequentially hybridized to nm23 and TIMP-1 probes in addition to GAPDH, as a control. Statistical analyses were carried out according to t-test for equality of means. RESULTS: NM23-H1 were detected in each sample, however it did not correlate with malignancy and invasiveness. On the other side TIMP-1 gene expression showed a clear correlation between low expression and invasiveness. CONCLUSION: The data suggest that TIMP-1 is an inhibitor of high grade gliomas invasion. NM23-H1 was present in the entire gliomas sample, but it did not vary in diffuse astrocytomas and glioblastomas.
Collapse
Affiliation(s)
- José Augusto Nasser
- Escola Paulista de Medicina, Universidade Federal de São Paulo, SP-Brazil, Universidade Estácio de Sá, Avenida Ataulfo de Paiva 135/914, 22440-032 Rio de Janeiro RJ, Brazil.
| | | | | | | | | |
Collapse
|
15
|
Caffo M, Caruso G, Germanò A, Galatioto S, Meli F, Tomasello F. CD68 and CR3/43 immunohistochemical expression in secretory meningiomas. Neurosurgery 2006; 57:551-7; discussion 551-7. [PMID: 16145535 DOI: 10.1227/01.neu.0000170440.32720.77] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Secretory meningiomas (SMs) are unusual benign meningiomas. SMs are highly vascularized lesions, with angiomatous features and a perivascular arrangement, and they are accompanied frequently by massive peritumoral edema. Microglia have been called the brain's immune system, although the specific role and prognostic significance of microglia remain uncertain. The objective of this study was to analyze the expression of CD68, a macrophage marker specific for resting microglia, and the expression of major histocompatibility complex Class II CR3/43 in SMs. METHODS From 1995 to 2002, six patients with SMs were treated surgically at our institution. Immunohistochemistry was used to analyze the expression of CD68 and CR3/43 in tumor specimens. The intensity of expression was graded semiquantitatively. A correlation between immunohistochemical expression and the occurrence of brain edema was studied. RESULTS CD68-positive mononuclear cells were observed in neoplastic tissue or around pseudopsammoma bodies and in perivascular areas, with minimal expression in one patient and moderate expression in three patients. CR3/43-positive complexes were detected in mononuclear elongated elements with ameboid extensions, presumably referable to cells at different stages of immunological activation phenomena, with minimal expression in two patients, moderate expression in one patient, and marked expression in one patient. Edema was severe in all patients. Therefore, it may be indirectly hypothesized that edema may not be correlated with the CD68 and CR3/43 immunohistochemical expression. CONCLUSION Macrophage infiltration and major histocompatibility complex Class II immunoreactivity in this subtype of meningioma suggest the occurrence of an immune response in the presence of SM.
Collapse
Affiliation(s)
- Mariella Caffo
- Neurosurgical Clinic, Department of Neurosciences, Psychiatry and Anaesthesiology, University of Messina School of Medicine, Messina, Italy
| | | | | | | | | | | |
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: 128] [Impact Index Per Article: 6.4] [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
|
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: 1.8] [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
|
18
|
Akasaki Y, Liu G, Chung NHC, Ehtesham M, Black KL, Yu JS. Induction of a CD4+ T Regulatory Type 1 Response by Cyclooxygenase-2-Overexpressing Glioma. THE JOURNAL OF IMMUNOLOGY 2004; 173:4352-9. [PMID: 15383564 DOI: 10.4049/jimmunol.173.7.4352] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PGE(2), synthesized by cyclooxygenase-2 (COX-2)-overexpressing tumor, is known to contribute to cellular immune suppression in cancer patients, but the mechanism remains unclear. We report the mechanism of a CD4(+) T regulatory type 1 (Tr1) induction by CD11c(+) mature dendritic cells (DCs) that phagocytose allogeneic and autologous COX-2-overexpressing glioma. A human glioma cell line, U-87MG, and primary cultured glioblastoma cells (MG-377) overexpressed COX-2. We did not detect IL-10Ralpha expression in these gliomas, and rIL-10 did not suppress their COX-2 expression. Exposure to COX-2-overexpressing glioma induced mature DCs to overexpress IL-10 and decreased IL-12p70 production. These DCs induced a Tr1 response, which is characterized by robust secretion of IL-10 and TGF-beta with negligible IL-4 secretion by CD4(+) T cells, and an inhibitory effect on admixed lymphocytes. Peripheral CD4(+) T cell populations isolated from an MG-377 patient also predominantly demonstrated a Tr1 response against MG-377 cells. Selective COX-2 inhibition in COX-2-overexpressing gliomas at the time of phagocytic uptake by DCs abrogated this regulatory response and instead elicited Th1 activity. COX-2 stable transfectants in LN-18 (LN-18-COX2) also induced a Tr1 response. The effect of a COX-2 inhibition in LN-18-COX2 is reversible after administration of PGE(2). Taken together, robust levels of PGE(2) from COX-2-overexpressing glioma, which is unresponsive to IL-10 within the local microenvironment, may cause DCs to secrete high levels of IL-10. These results indicate that COX-2-overexpressing tumors induce a Tr1 response, which is mediated by tumor-exposed, IL-10-enhanced DCs.
Collapse
Affiliation(s)
- Yasuharu Akasaki
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | | |
Collapse
|
19
|
Walker PR, Calzascia T, de Tribolet N, Dietrich PY. T-cell immune responses in the brain and their relevance for cerebral malignancies. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2003; 42:97-122. [PMID: 12738053 DOI: 10.1016/s0165-0173(03)00141-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In order that cellular immune responses afford protection without risk to sensitive normal tissue, they must be adapted to individual tissues of the body. Nowhere is this more critical than for the brain, where various passive and active mechanisms maintain a state of immune privilege that can limit high magnitude immune responses. Nevertheless, it is now clear that immune responses are induced to antigens in the brain, including those expressed by cerebral malignancies. We discuss hypotheses of how this can occur, although details such as which antigen presenting cells are involved remain to be clarified. Antitumor responses induced spontaneously are insufficient to eradicate malignant astrocytomas; many studies suggest that this can be explained by a combination of low level immune response induction and tumor mediated immunosuppression. A clinical objective currently pursued is to use immunotherapy to ameliorate antitumour immunity. This will necessitate a high level immune response to ensure sufficient effector cells reach the tumor bed, focused cytotoxicity to eradicate malignant cells with little collateral damage to critical normal cells, and minimal inflammation. To achieve these aims, priority should be given to identifying more target antigens in astrocytoma and defining those cells present in the brain parenchyma that are essential to maintain antitumour effector function without exacerbating inflammation. If we are armed with better understanding of immune interactions with brain tumor cells, we can realistically envisage that immunotherapy will one day offer hope to patients with currently untreatable neoplastic diseases of the CNS.
Collapse
Affiliation(s)
- Paul R Walker
- Laboratory of Tumour Immunology, Division of Oncology, Geneva University Hospital, 24 rue Micheli-du-Crest, 1211 Geneva 14, Switzerland.
| | | | | | | |
Collapse
|
20
|
Abstract
Microglia have long been ignored by neurooncologists. This has changed with the realization that microglial cells not only occur within and around brain tumors but also contribute significantly to the actual tumor mass, notably in astrocytic gliomas. In addition, it has been speculated that microglia could play a role in the defense against neoplasms of the nervous system. However, the biological success of these tumors, i.e., their highly malignant behavior, indicates that natural microglial defense mechanisms do not function properly in astrocytomas. In fact, there is evidence that microglial behavior is controlled by tumor cells, supporting their growth and infiltration. This unexpected "Achilles heel" of microglial immune defense illustrates the risk of generalizing on the basis of a single aspect of microglial biology. Microglia are highly plastic cells, capable of exerting cytotoxic functions under conditions of CNS infections, but not necessarily during glioma progression. Thus, the suggestion that microglial activation through stimulation by cytokines (e.g., interferon-gamma) will benefit patients with brain tumors could prove fatally wrong. Therapeutic recruitment of microglia to treat such diffusely infiltrative brain tumors as astrocytic gliomas must be considered premature.
Collapse
Affiliation(s)
- Manuel B Graeber
- Department of Neuropathology, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Bernd W Scheithauer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Georg W Kreutzberg
- Department of Neuromorphology, Max-Planck-Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
21
|
Subramanian A, Harris A, Piggott K, Shieff C, Bradford R. Metastasis to and from the central nervous system--the 'relatively protected site'. Lancet Oncol 2002; 3:498-507. [PMID: 12147436 DOI: 10.1016/s1470-2045(02)00819-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The brain has long been recognised as a site with a very low rate of metastases, despite the potential for cancers to be extremely locally aggressive. This feature contrasts with most of the rest of the body, where metastatic spread is much more common. The pathological behaviour of any tumour is governed by both its inherent composition and the composition of the matrix in which it is sited. Much work has been done in recent years to elucidate the factors within the central nervous system (CNS) that give the brain its unique properties. Tumour interactions with the blood-brain barrier, microglia, and various matrix proteins, cytokines, and growth factors have a central role. This review concentrates mainly on the process of tumour spread from the CNS and explores how the brain is a protected site. CNS metastases from extraneural sites are also briefly covered.
Collapse
Affiliation(s)
- A Subramanian
- Department of Neurosurgery, Royal Free Hospital and School of Medicine, London, UK.
| | | | | | | | | |
Collapse
|
22
|
Cuny E, Loiseau H, Penchet G, Ellie E, Arsaut J, Vital A, Vincendeau P, Demotes-Mainard J. Association of elevated glial expression of interleukin-1beta with improved survival in patients with glioblastomas multiforme. J Neurosurg 2002; 96:294-301. [PMID: 11838804 DOI: 10.3171/jns.2002.96.2.0294] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The aim of this study was to investigate the association of interleukin-1beta (IL-1beta) expression with improved survival in patients with glioblastomas multiforme (GBMs). Immune and vascular host-tumor interactions play a pivotal role in the control of tumor development, and inflammatory mechanisms may participate in the host's defense against tumor cells. Expression of proinflammatory cytokines and of inducible nitric oxide synthase (iNOS) has been noted in various types of malignant tumors, raising the possibility that endogenous expression of cytokines and the resulting cytotoxic action of sustained NO production play a role in the control of tumor growth. Indeed, human GBMs express variable amounts of iNOS. METHODS In this study, the expression of iNOS and of cytokines known to upregulate IL-1beta, tumor necrosis factor-alpha, interferon-gamma or downregulate iNOS transcription (IL-10, transforming growth factor [TGF]beta1, and TGFbeta2) were measured using reverse transcription-polymerase chain reaction with competitor DNA in 39 samples of human GBM. The iNOS level in GBM was positively correlated with IL-1beta messenger (m)RNA, but not with the other cytokines tested. Immunocytochemical double labeling revealed that both anti-iNOS immunoreactivity and anti-IL-1beta immunoreactivity colocalized with glial fibrillary acidic protein immunoreactivity in GBM. Some macrophage/microglial cells also expressed iNOS, but not IL-1beta. Comparison of biological data with clinical parameters indicated that the survival duration was enhanced when levels of IL-1beta mRNA were elevated or when levels of TGFbeta2 were low, but was independent of the level of iNOS mRNA within the tumor. CONCLUSIONS Taken together, these data indicate that the proinflammatory cytokine IL-1beta produced within GBM by glial-derived cells has a negative impact on tumor growth through a mechanism independent of iNOS induction.
Collapse
Affiliation(s)
- Emmanuel Cuny
- INSERM U-394 Neurobiologie Intégrative, Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Sasaki A, Ishiuchi S, Kanda T, Hasegawa M, Nakazato Y. Analysis of interleukin-6 gene expression in primary human gliomas, glioblastoma xenografts, and glioblastoma cell lines. Brain Tumor Pathol 2002; 18:13-21. [PMID: 11517969 DOI: 10.1007/bf02478920] [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: 02/05/2023]
Abstract
Our previous study showed that high-grade astrocytomas often expressed high interleukin (IL)-1beta production. Coexpression of IL-1beta and IL-6 has been found in a number of glioma samples and glioma cell lines. To characterize the expression of IL-6 in the human glioma microenvironment, we investigated surgically excised human gliomas, human glioblastoma xenografts, and human glioblastoma cell lines using the reverse transcriptase-polymerase chain reaction (RT-PCR), immunohistochemistry (IHC), and enzyme-linked immunosorbent assay (ELISA). In the 29 primary gliomas, transcripts of IL-6 were less frequently detectable (55.6%) than those of IL-1beta (72.4%) or those of IL-10, IL-8, or IL-1alpha (>80% each). As for IL-6 gene expression, little or no transcription was observed in low-grade astrocytomas, oligodendroglial tumors, and 1 ependymoma. Strong IL-6 gene expression was found in only 5 of 9 glioblastomas. Immunohistochemically, IL-6 antigen was localized in the tumor cells and macrophages in 4 of 7 glioblastomas. In 3 glioblastomas transplanted into nude mice, both IL-1beta and IL-6 were detected only in 1, but othercytokines (IL-8, IL-10, and IL-1alpha) were detected in all 3 xenografts by RT-PCR. Two cell lines both showed IL-6 expression at the mRNA level, and in a cell line with a high level of IL-6 and IL-1beta transcripts, significant production of IL-6 was observed by IHC and ELISA. We concluded that IL-6 produced in tumor tissue may be involved in tumor progression in some glioblastomas, but not in low-grade astrocytomas and oligodendroglial tumors, and that IL-6 gene expression is closely correlated with IL-1beta expression in biopsy tissue, xenografts, and cultures of human gliomas.
Collapse
Affiliation(s)
- A Sasaki
- Department of Pathology, Gunma University School of Medicine, Maebashi, Japan.
| | | | | | | | | |
Collapse
|
24
|
Affiliation(s)
- P R Walker
- Laboratory of Tumor Immunology, Division of Oncology, University Hospital, Rue Micheli-du-Crest 24, 1211 Geneva 14, Switzerland.
| | | |
Collapse
|
25
|
Abstract
BACKGROUND The incidence of brain tumors is increasing rapidly, particularly in the older population. Advances in molecular biology help to explain differences in biologic behavior and response to therapy of brain tumors in the elderly compared with younger patients. The number of elderly patients who desire and receive therapy for brain tumors and are included in clinical trials is increasing. METHODS This article reviews the literature on the epidemiology, clinical aspects, and therapy of brain tumors, with emphasis on the older patient population. RESULTS The increased incidence of brain tumors in the elderly is principally due to the increasing number of people who comprise the older population. Age and performance status are important independent prognostic indicators, together with tumor histology. Surgery, radiation therapy, and chemotherapy can benefit elderly patients with brain tumors with favorable histologies, tumor location, and good performance status. The response rates to available therapies are less favorable than in younger patients, and only a small number of elderly patients are enrolled in clinical studies addressing new treatment modalities. CONCLUSIONS Brain tumors in the elderly have specific characteristics that determine their biologic behavior and response to therapy. There is a need for clinical studies designed for treatment of brain tumors in older patients, and requirements for rehabilitation and support systems for the elderly need to be addressed.
Collapse
Affiliation(s)
- A Flowers
- Department of Neurology, Hartford Hospital, CT 06102-5037, USA.
| |
Collapse
|
26
|
Taniguchi Y, Ono K, Yoshida S, Tanaka R. Antigen-presenting capability of glial cells under glioma-harboring conditions and the effect of glioma-derived factors on antigen presentation. J Neuroimmunol 2000; 111:177-85. [PMID: 11063836 DOI: 10.1016/s0165-5728(00)00361-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The antigen-presenting capability of syngeneic rat glial cells was investigated under glioma-harboring conditions. Microglia induced a significant proliferation of glioma-primed splenocytes, but astrocytes did not. Furthermore, astrocytes suppressed the accessory cell function of microglia. The presence of both indomethacin and anti-interleukin (IL)-10 neutralizing antibody during priming of microglia enhanced splenocyte proliferation. The glioma culture supernatants down-regulated the interferon-gamma-induced expression of major histocompatibility complex class II molecules on microglia. The down-regulation was blocked by indomethacin and anti-IL-10 antibody. The results suggest that microglia but not astrocytes may function as antigen-presenting cells in glioma, and that glioma may suppress the antigen-presenting abilities of microglia.
Collapse
Affiliation(s)
- Y Taniguchi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Asahimachi 1, 951-8585, Niigata, Japan
| | | | | | | |
Collapse
|
27
|
Abstract
Recent studies suggest that neurocysticercosis may be a risk factor for human cancer. Pathogenetic mechanisms explaining possible oncogenic effects of cysticerci include the following: (a) parasite-induced modulation of the host immune response that may be associated with loss of regulatory mechanisms implicated in the immunological surveillance against cancer; (b) transfer of genetic material from the parasite to the host, causing DNA damage and malignant transformation of host cells, and (c) chronic inflammation with liberation of nitric oxide and inhibition of tumor suppressor genes. Further research is needed to confirm the potential role of cysticercosis in the development of cancer. These studies should determine the presence of cysticercotic factors responsible for the transfer of genetic material and potential mutations in the tumor suppressor genes in proliferating astrocytes surrounding cysticercotic lesions. Additionally, the complex interaction between the immune state of the host with variable cytokine release and the presence of inflammatory cells releasing nitric oxide that cause DNA damage and impair tumor suppressive mechanisms needs to be investigated.
Collapse
Affiliation(s)
- O H Del Brutto
- Departmento de Neurología, Hospital Luis Vernaza, Guayaquil, Ecuador.
| | | | | | | |
Collapse
|
28
|
Affiliation(s)
- J Sotelo
- División de Investigación, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, México, D.F., Mexico.
| | | |
Collapse
|
29
|
Dix AR, Brooks WH, Roszman TL, Morford LA. Immune defects observed in patients with primary malignant brain tumors. J Neuroimmunol 1999; 100:216-32. [PMID: 10695732 DOI: 10.1016/s0165-5728(99)00203-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Malignant glioblastomas (gliomas) account for approximately one third of all diagnosed brain tumors. Yet, a decade of research has made little progress in advancing the treatment of these tumors. In part this lack of progress is linked to the challenge of discovering how glial tumors are capable of both modulating host immune function and neutralizing immune-based therapies. Patients with gliomas exhibit a broad suppression of cell-mediated immunity. The impaired cell-mediated immunity observed in patients with gliomas appears to result from immunosuppressive factor(s) secreted by the tumor. This article reviews what has been elucidated about the immune defects of patients harboring glioma and the glioma-derived factors which mediate this immunosuppression. A model involving systemic cytokine dysregulation is presented to suggest how the immune defects arise in these individuals.
Collapse
Affiliation(s)
- A R Dix
- Department of Microbiology and Immunology, University of Kentucky Medical Center, Lexington 40536-0084, USA
| | | | | | | |
Collapse
|
30
|
Asai J, Suzuki R, Fujimoto T, Suzuki T, Nakagawa N, Nagashima G, Miyo T, Hokaku H, Takei A. Fluorescence automatic cell sorter and immunohistochemical investigation of CD68-positive cells in meningioma. Clin Neurol Neurosurg 1999; 101:229-34. [PMID: 10622450 DOI: 10.1016/s0303-8467(99)00052-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Infiltration of brain neoplasms by mononuclear cells including monocytes/macrophages has attracted little attention since they have marked morphological heterogeneity. Twenty-seven meningiomas were studied by anti-CD68 antibody-gated flow cytometry and by immunohistochemical analysis using the anti-CD68 antibodies. Flow cytometric analysis divided cells contained within tumor tissues into CD68-positive and -negative cells. In addition, eight gliomas, eight metastatic brain tumor, and 12 pituitary adenomas were investigated in the same way to compare meningiomas. The mean contents of CD68-positive cells were 24.0 +/- 3.7% in meningiomas, 4.4 +/- 1.4% in gliomas, 9.5 +/- 3.9% in metastatic brain tumors, and 4.5 +/- 1.8% in pituitary adenomas. Immunohistochemically, CD68-positive cells showed significant heterogeneity and were detected as round, rod-shaped, ameboid and ramified cells in meningiomas. Although the infiltrated mononuclear cells in gliomas have been investigated to some degree and showed that they express cytokines and/or growth factors, these infiltrated cells in meningioma have barely been studied. The CD68-positive cells detected in this study are likely to be monocytes, macrophages and microglias, and are presumed to be in various functional stages and to play important roles in growth regulation in meningioma.
Collapse
Affiliation(s)
- J Asai
- Department of Neurosurgery, Fujigaoka Hospital, Showa University, Yokohama, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Liau LM, Black KL, Prins RM, Sykes SN, DiPatre PL, Cloughesy TF, Becker DP, Bronstein JM. Treatment of intracranial gliomas with bone marrow-derived dendritic cells pulsed with tumor antigens. J Neurosurg 1999; 90:1115-24. [PMID: 10350260 DOI: 10.3171/jns.1999.90.6.1115] [Citation(s) in RCA: 184] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT An approach toward the treatment of intracranial gliomas was developed in a rat experimental model. The authors investigated the ability of "professional" antigen-presenting cells (dendritic cells) to enhance host antitumor immune responses when injected as a vaccine into tumor-bearing animals. METHODS Dendritic cells, the most potent antigen-presenting cells in the body, were isolated from rat bone marrow precursors stimulated in vitro with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4. Cultured cell populations were confirmed to be functional antigen-presenting cells on the basis of expressed major histocompatibility molecules, as analyzed by fluorescence-activated cell sorter cytofluorography. These dendritic cells were then pulsed (cocultured) ex vivo with acid-eluted tumor antigens from 9L glioma cells. Thirty-eight adult female Fischer 344 rats harboring 7-day-old intracranial 9L tumors were treated with three weekly subcutaneous injections of either control media (10 animals), unpulsed dendritic cells (six animals), dendritic cells pulsed with peptides extracted from normal rat astrocytes (10 animals), or 9L tumor antigen-pulsed dendritic cells (12 animals). The animals were followed for survival. At necropsy, the rat brains were removed and examined histologically, and spleens were harvested for cell-mediated cytotoxicity assays. The results indicate that tumor peptide-pulsed dendritic cell therapy led to prolonged survival in rats with established intracranial 9L tumors implanted 7 days prior to the initiation of vaccine therapy in vivo. Immunohistochemical analyses were used to document a significantly increased perilesional and intratumoral infiltration of CD8+ and CD4+ T cells in the groups treated with tumor antigen-pulsed dendritic cells compared with the control groups. In addition, the results of in vitro cytotoxicity assays suggest that vaccination with these peptide-pulsed dendritic cells can induce specific cytotoxic T lymphocytes against 9L tumor cells. CONCLUSIONS Based on these results, dendritic antigen-presenting cells pulsed with acid-eluted peptides derived from autologous tumors represent a promising approach to the immunotherapy of established intracranial gliomas. which may serve as a basis for designing clinical trials in patients with brain tumors.
Collapse
Affiliation(s)
- L M Liau
- Department of Neurology, Jonsson Comprehensive Cancer Center, University of California at Los Angeles School of Medicine, 90095, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Damage to the central nervous system (CNS) elicits the activation of both astrocytes and microglia. This review is focused on the principal features that characterize the activation of microglia after CNS injury. It provides a critical discussion of concepts regarding microglial biology that include the relationship between microglia and macrophages, as well as the role of microglia as immunocompetent cells of the CNS. Mechanistic and functional aspects of microgliosis are discussed primarily in the context of microglial neuronal interactions. The controversial issue of whether reactive microgliosis is a beneficial or a harmful process is addressed, and a resolution of this dilemma is offered by suggesting different interpretations of the term 'activated microglia' depending on its usage during in vivo or in vitro experimentation.
Collapse
Affiliation(s)
- W J Streit
- Department of Neuroscience, University of Florida College of Medicine and Brain Institute, Gainesville 32610, USA.
| | | | | |
Collapse
|
33
|
Sultana S, Zhou R, Sadagopan MS, Skalli O. Effects of growth factors and basement membrane proteins on the phenotype of U-373 MG glioblastoma cells as determined by the expression of intermediate filament proteins. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 153:1157-68. [PMID: 9777947 PMCID: PMC1853038 DOI: 10.1016/s0002-9440(10)65660-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Various growth factors and basement membrane proteins have been implicated in the pathobiology of astrocytomas. The goal of this study was to determine the relative contribution of these two factors in modulating the phenotype of U-373 MG glioblastoma cells as determined by the expression of the intermediate filament proteins glial fibrillary acidic protein, vimentin, and nestin. For these determinations, cells plated in serum-free medium were treated either with growth factors binding to tyrosine kinase receptors including transforming growth factor-alpha, epidermal growth factor, platelet-derived growth factor-AA, basic fibroblast growth factor, and insulin-like growth factor-1 or with basement membrane proteins including collagen IV, laminin, and fibronectin. The changes in the expression levels of intermediate filament proteins in response to these treatments were analyzed by quantitation of immunoblots. The results demonstrate that collagen IV and growth factors binding to tyrosine kinase receptors decrease the glial fibrillary acidic protein content of U-373 MG cells. Growth factors binding to tyrosine kinase receptors also decrease the vimentin content of these cells but do not affect their nestin content. On the other hand, basement membrane proteins decrease the nestin content of U-373 MG cells but do not affect their vimentin content. The significance of these results with respect to the role played by different factors in modulating the phenotype of neoplastic astrocytes during tumor progression is discussed.
Collapse
Affiliation(s)
- S Sultana
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 60612, USA
| | | | | | | |
Collapse
|
34
|
Moreland LW. Soluble tumor necrosis factor receptor (p75) fusion protein (ENBREL) as a therapy for rheumatoid arthritis. Rheum Dis Clin North Am 1998; 24:579-91. [PMID: 9710888 DOI: 10.1016/s0889-857x(05)70027-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Soluble tumor necrosis factor (TNF) receptor fusion protein (p75) (Enbrel) is a reversible inhibitor of the biologic effects of TNF. Enbrel has been shown in placebo-controlled trials to significantly improve the signs and symptoms of rheumatoid arthritis. Clinical trials are now in progress to assess the safety and efficacy of Enbrel in combination with methotrexate in refractory rheumatoid arthritis along with trials to compare Enbrel to methotrexate in patients with early rheumatoid arthritis.
Collapse
Affiliation(s)
- L W Moreland
- Department of Medicine, University of Alabama at Birmingham, USA.
| |
Collapse
|
35
|
Tran CT, Wolz P, Egensperger R, Kösel S, Imai Y, Bise K, Kohsaka S, Mehraein P, Graeber MB. Differential expression of MHC class II molecules by microglia and neoplastic astroglia: relevance for the escape of astrocytoma cells from immune surveillance. Neuropathol Appl Neurobiol 1998; 24:293-301. [PMID: 9775395 DOI: 10.1046/j.1365-2990.1998.00120.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is increasing evidence that microglia serve as antigen presenters in the human CNS. Although the occurrence of MHC class II immunoreactive cells has been reported in astrocytic gliomas, the relative contribution of microglia to this cell population has not been studied in detail. Using computer-assisted image analysis, we have investigated the expression of MHC class II molecules and of the microglia/macrophage markers Ki-MIP, RCA-1, KP1 and iba1, in 97 astrocytic gliomas comprising all WHO grades to answer the question whether there is a correlation between tumour grade and the number of MHC class II positive microglia/macrophage profiles. Microglia expressing MHC class II were common in astrocytomas and anaplastic astrocytomas but rare in pilocytic tumours although there was significant variation within each group. MHC class II immunoreactivity was reduced in highly cellular areas of glioblastomas where large numbers of cells expressing macrophage markers were still present. Thus, there was no simple relationship between tumour grade and microglial/macrophage MHC class II expression. In addition, up to 55% of astrocytic gliomas contained MHC class II immunoreactive tumour cells. Microglia but not tumour cells were found to express the BB1/B7 costimulator. We conclude that microglia in astrocytic gliomas are well equipped to function as antigen presenting cells. Yet, neoplastic astroglia appear to acquire the capacity to downregulate microglial MHC class II expression and, at the same time, may induce T-cell clonal anergy through aberrant expression of MHC class II molecules.
Collapse
Affiliation(s)
- C T Tran
- Institute of Neuropathology, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Esteve PO, Tremblay P, Houde M, St-Pierre Y, Mandeville R. In vitro expression of MMP-2 and MMP-9 in glioma cells following exposure to inflammatory mediators. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1403:85-96. [PMID: 9622599 DOI: 10.1016/s0167-4889(98)00020-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Progression of glioma is associated with local degenerative processes which are attributed to the activity of gelatinases. As glioma cells are candidate for secretion of these enzymes, we have studied in vitro the potential of cytokines (interleukin-1alpha (IL-1), tumor necrosis factor-alpha (TNFalpha) and transforming growth factor-beta (TGFbeta2)) to regulate the expression of gelatinase A and B (Gels A and B, respectively) in two glioma cells of human (A172) and rat origin (C6). We showed that IL-1 and TNFalpha both induced gene expression and protein secretion of Gel B in both cell lines, as revealed by RT-PCR and gelatin zymography, respectively. In C6 cells, TNFalpha had no effect on Gel A constitutive expression while IL-1 increased its production, but only at high doses. We have also demonstrated that TGFbeta2 inhibited both IL-1- or TNFalpha-induced gene expression and Gel B production in a dose-dependent manner but had no effect on Gel A secretion. The effect of TGFbeta2 on Gel B secretion was reversed by phorbol myristate acetate (PMA). Taken together, these data suggest that IL-1, TNFalpha and TGFbeta2 tightly regulate Gel B secretion in glioma cells, an enzyme which is believed to play an important role in the local invasion of brain tissue by tumor cells.
Collapse
Affiliation(s)
- P O Esteve
- Institut Armand-Frappier, Centre de Recherches en Immunologie, CP 100, Laval, Quebec, Canada
| | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- L Lorusso
- Department of Pathology, Walton Hospital, Liverpool, United Kingdom
| | | |
Collapse
|
38
|
Zlokovic BV, Apuzzo ML. Cellular and molecular neurosurgery: pathways from concept to reality--part I: target disorders and concept approaches to gene therapy of the central nervous system. Neurosurgery 1997; 40:789-803; discussion 803-4. [PMID: 9092853 DOI: 10.1097/00006123-199704000-00027] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent advances in cellular and molecular biology and better understanding of genetic and biochemical bases of different central nervous system (CNS) disorders have made gene therapy of the CNS a realistic goal. Concept approaches for gene therapy of CNS disorders are reviewed and include the following: 1) gene replacement with a single normal allele to correct the inherited global neurodegenerative disorders, such as enzyme deficiencies; 2) brain repair to restore the function of a particular subset of cells that were lost because of a neurodegenerative process; 3) gene therapy of brain tumors; and 4) gene therapy of stroke. Techniques of viral vector-mediated CNS transfer of a therapeutic gene, transplantation of genetically modified cells, fetal embryonic implantation and/or implantation of genetically engineered neural progenitor cells, and production of a specific enzyme, neurotransmitter, and/or growth factor are discussed with respect to the therapeutic potential for global and localized CNS neurodegenerative disorders and stroke. Transfection of the CNS tumor cells with the drug susceptibility ("suicide") gene and/or "toxic" gene and antisense strategies and a concept of adoptive immunotherapy of brain tumors are also discussed. Other approaches, such as transfer of drug-resistant genes and monoclonal antibody gene transfer, are briefly discussed. In addition to summarizing current principles of gene therapy for several groups of CNS disorders, the issues that remain to be resolved in clinical reality, such as delivery of the genetic material and regulation of the cellular expression of the transgene, and the negatives associated with the concepts of gene therapy, such as transient gene expression, toxicity of viral proteins, drawbacks of antisense therapy, and the problem of immune response to the transfected protein, have been also identified.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Brain Diseases, Metabolic/genetics
- Brain Diseases, Metabolic/surgery
- Brain Diseases, Metabolic/therapy
- Brain Neoplasms/genetics
- Brain Neoplasms/immunology
- Brain Neoplasms/surgery
- Brain Neoplasms/therapy
- Brain Tissue Transplantation
- Cell Transplantation
- Central Nervous System Diseases/genetics
- Central Nervous System Diseases/surgery
- Central Nervous System Diseases/therapy
- Cerebrovascular Disorders/genetics
- Cerebrovascular Disorders/therapy
- DNA, Recombinant/genetics
- DNA, Recombinant/therapeutic use
- Drug Resistance/genetics
- Enzymes/deficiency
- Enzymes/genetics
- Fetal Tissue Transplantation
- Genetic Diseases, Inborn/surgery
- Genetic Diseases, Inborn/therapy
- Genetic Therapy/methods
- Genetic Vectors
- Humans
- Immunotherapy, Adoptive
- Mice
- Mice, Knockout
- Neurosurgery/methods
- Neurosurgery/trends
- Oligonucleotides, Antisense/therapeutic use
- Parkinson Disease/surgery
- Stem Cell Transplantation
- Transfection
- Transgenes
Collapse
Affiliation(s)
- B V Zlokovic
- Department of Neurological Surgery, Childrens Hospital Los Angeles, University of Southern California School of Medicine, USA
| | | |
Collapse
|
39
|
Chen TC, Hinton DR, Yong VW, Hofman FM. TGF-B2 and soluble p55 TNFR modulate VCAM-1 expression in glioma cells and brain derived endothelial cells. J Neuroimmunol 1997; 73:155-61. [PMID: 9058771 DOI: 10.1016/s0165-5728(96)00190-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Transforming growth factor beta-2 (TGF-B2) is secreted by glioma cells and is known to decrease leukocyte-endothelium interaction. TGF-B2 alone and in conjunction with soluble tumor necrosis factor (TNF) p55 receptor, was found to decrease the expression of TNF induced VCAM-1 on the malignant glioma cell line A-172 and human cerebral microvessel endothelial (CNS-EC) cells. Co-culture of A-172 glioma cells led to a decrease in VCAM-1 expression; this effect on CNS-EC in co-culture could be simulated by glioma supernatant alone. These results suggest that malignant gliomas, by secreting TGF-B2 and releasing soluble TNF receptors, modulate adhesion molecules.
Collapse
Affiliation(s)
- T C Chen
- Department of Pathology, School of Medicine, University of Southern California, Los Angeles 90033, USA
| | | | | | | |
Collapse
|
40
|
Baudet C, Chevalier G, Chassevent A, Canova C, Filmon R, Larra F, Brachet P, Wion D. 1,25-Dihydroxyvitamin D3 induces programmed cell death in a rat glioma cell line. J Neurosci Res 1996; 46:540-50. [PMID: 8951666 DOI: 10.1002/(sici)1097-4547(19961201)46:5<540::aid-jnr3>3.0.co;2-j] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
1,25-Dihydroxyvitamin D3 (1,25(OH)2D3), a seco-steroid hormone with potential antitumoral activities, has been recently reported to exert cytotoxic effects on C6 glioma cells. However, the molecular mechanisms which trigger this cell death remain unknown. We show here that this 1,25(OH)2D3-induced cell death is dependent upon protein synthesis and is accompanied by the expression of c-myc, p53, and gadd45 genes. Two other genes, coding for interleukin-6 and vaso-endothelial growth factor, are also upregulated after addition of 1,25(OH)2D3. This programmed cell death can be suppressed when cells are treated with forskolin, a drug which increases intracellular cAMP concentration, or with genistein, an inhibitor of tyrosine protein kinases. However, in spite of the demonstration of fragmented DNA in 1,25(OH)2D3-treated cells, the C6.9 cells used in this study do not show the classical morphological features of apoptosis. These results provide the first evidence for the existence of a programmed cell death triggered by 1,25(OH)2D3 in glioma cells and may provide a basis for the development of new therapeutic strategies. In addition, these data also suggest that the treatment of C6.9 cells with 1,25(OH)2D3 may be a useful model to study the molecular mechanisms involved in the programmed cell death of a cell of glial origin.
Collapse
Affiliation(s)
- C Baudet
- INSERM U 298, Centre Hospitalier Universitaire, Angers, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Fries G, Perneczky A, Kempski O. Glioblastoma-associated circulating monocytes and the release of epidermal growth factor. J Neurosurg 1996; 85:642-7. [PMID: 8814168 DOI: 10.3171/jns.1996.85.4.0642] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Monocytes/macrophages frequently infiltrate malignant gliomas and play a central role in the tumor-associated immune response as they process tumor antigen and present it to T-lymphocytes. Findings have accumulated that peripheral blood monocytes leaving the cerebral circulation become microglial cells and vice versa and that monocytes/macrophages may stimulate malignant tumor growth by some unknown mechanism. Most malignant gliomas express growth factor receptors, for example epidermal growth factor receptor (EGFR). The aim of this study was to determine whether peripheral blood monocytes of glioma patients release EGF, the appropriate ligand of gliomacell membrane-bound EGFR. Long-term cultured peripheral blood monocytes from 14 patients with malignant gliomas were compared to those from 12 controls (seven with nontumorous disease and five healthy individuals). Using an enzyme-linked immunosorbent assay for EGF, the EGF content of cell culture supernatants was determined at Days 7, 21, and 100 of culture. The EGF content (mean +/- standard error) of supernatants was 5.9 +/- 0.2 pg/ml/10(3) glioma monocytes versus 1.3 +/- 0.1 pg/ml/10(3) control monocytes at Day 7 of culture, 22.9 +/- 0.8 pg/ml/10(3) glioma monocytes versus 1.8 +/- 0.9 pg/ml/10(3) control monocytes at Day 21 of culture, and 23.4 +/- 0.7 pg/ml/10(3) glioma monocytes, and below detection levels for control monocytes at Day 100 of culture. Steroid treatment of glioma patients did not influence the EGF release of cultured monocytes. These data indicate that glioblastoma-associated peripheral blood monocytes may be distinct from those of healthy individuals. Moreover, this study indicates that subtypes of glioma-associated peripheral blood monocytes may support immunosuppression and promote growth of malignant glioma by releasing unusually high amounts of EGF.
Collapse
Affiliation(s)
- G Fries
- Department of Neurosurgery and Institute for Neurosurgical Pathophysiology, Johannes Gutenberg-University Medical School, Mainz, Germany
| | | | | |
Collapse
|
42
|
Levy EI, Paino JE, Sarin PS, Goldstein AL, Caputy AJ, Wright DC, Sekhar LN. Enzyme-linked immunosorbent assay quantification of cytokine concentrations in human meningiomas. Neurosurgery 1996; 39:823-8; discussion 828-9. [PMID: 8880778 DOI: 10.1097/00006123-199610000-00036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To gain insight into the network of cytokine gene expression in the brain tumor microenvironment, we investigated the presence of the following cytokines in freshly excised brain tumors: interleukin (IL)-1 beta, IL-2, IL-4, and IL-6. METHODS Tumor specimens from nine meningiomas were grown as tissue explants. The supernatants from the explants were tested for the presence of the aforementioned cytokines via the enzyme-linked immunosorbent assay method. RESULTS IL-6, which is thought to stimulate acute protein phase synthesis, neovascularization, and cell proliferation, was found in all of the samples in greater concentrations than the other cytokines tested. IL-1 beta, another stimulatory cytokine thought to be involved in acute protein phase synthesis and cell proliferation, was also found in 100% of the samples tested, in concentrations significantly lower than those of IL-6. As expected, the presence of IL-2 and IL-4 was not detectable in any of the samples. CONCLUSION This study is the first to clearly determine the relative concentrations of IL-1 beta and IL-6, using enzyme-linked immunosorbent assay quantification. These findings are an important precursor to future studies using antibodies to IL-1 beta and IL-6 and antibodies to IL-6 receptors to modulate neoplastic growth both in vitro and in vivo.
Collapse
Affiliation(s)
- E I Levy
- School of Medicine, George Washington University, District of Columbia, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Nam M, Johnston P, Lal B, Indurti R, Wilson MA, Laterra J. Endothelial cell-based cytokine gene delivery inhibits 9L glioma growth in vivo. Brain Res 1996; 731:161-70. [PMID: 8883866 DOI: 10.1016/0006-8993(96)00471-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Malignant brain neoplasms present great therapeutic challenges due to their extremely aggressive behavior and relative isolation by the blood-brain and blood-tumor barriers. Endothelial cells may be versatile platforms for delivering genes to solid tumors by virtue of their location at blood-tissue interfaces and their proliferation in response to endothelial mitogens produced by tumors. Immortalized rat brain endothelial cells that express the E. coli lacZ reporter gene and the gene for murine interleukin-2 (RBEZ-IL2) were co-inoculated with 9L glioma cells to Fisher rats to examine the effects of endothelial cell-based cytokine delivery on glioma growth in vivo. 9L glioma growth was not affected by the implantation of control RBEZ cells. The growth of subcutaneous and intracranial 9L gliomas was significantly inhibited by RBEZ-IL2 cells (P < 0.005 and P < 0.01, respectively) when compared to control transfected RBEZ cells. Rats receiving intracranial 9L glioma cells with RBEZ-IL2 cells showed increased survival (P < 0.001). Histologic and immunohistologic analysis showed enhanced activation of microglia/macrophages and CD8-positive T lymphocytes and/or natural killer cells within brain at sites of 9L inoculation with RBEZ-IL2 cells. This report establishes that immortalized endothelial cells can be used for cytokine gene delivery and to activate anti-tumor host responses to experimental gliomas within the central nervous system.
Collapse
Affiliation(s)
- M Nam
- Department of Neurology, Kennedy Krieger Research Institute, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
44
|
Herrlinger U, Buchholz R, Jachimczak P, Schabet M. Intrathecal treatment of C6 glioma leptomeningeal metastasis in Wistar rats with interleukin-2. J Neurooncol 1996; 27:193-203. [PMID: 8847552 DOI: 10.1007/bf00165475] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The efficacy of intrathecal treatment of leptomeningeal metastasis (LM) with interleukin-2 (IL-2) was evaluated in an animal model using Wistar rats inoculated intracisternally with 10(7) C6 glioma cells. Prior to the in vivo experiments the antiproliferative effects of human IL-2, and of murine IFN-gamma and TNF-alpha which are cytokines induced by IL-2 were tested in a colony forming assay. Only IFN-gamma caused a dose-dependent inhibition of colony formation. Twelve animals were treated intracisternally with either 10(5) IU IL-2 or control medium on day 0, 2, and 5 after tumor cell inoculation. Both IL-2 treated and sham-treated animals developed LM with a symptom-free survival of 7 to 9 days. There was no significant difference between treated and untreated animals regarding time to onset of symptoms and pattern of tumor growth. Infiltration of the tumor tissue with ED-1+ monocytes and macrophages, and CD8+ lymphocytes, however, was slightly increased in IL-2 treated animals. In a second experiment 4 non tumor-bearing Wistar rats were intracisternally injected with a single dose of 10(5) IU IL-2. These animals also showed slightly enhanced leptomeningeal infiltration with CD8+ lymphocytes compared to controls. We conclude that intrathecal application of high-dose IL-2 although eliciting a slight immune reaction within the leptomeninges does not inhibit leptomeningeal tumor growth or prolong symptom-free survival in our animal model of LM. These results raise doubt about the clinical efficacy of intrathecal IL-2 treatment in patients with LM.
Collapse
Affiliation(s)
- U Herrlinger
- Department of Neurology, University of Tuebingen, Germany
| | | | | | | |
Collapse
|
45
|
Abstract
Effectively, modern research has confirmed Hortega's view of the origin of the microgliacyte from circulating monocytes of the monocyte-macrophage series that invade the brain during embryonic and early postnatal life. Their phagocytic capacity is exercised during the brain remodelling that marks brain maturation. They then convert to the ramified resting microglial cell visualized in the silver carbonate staining technique of Hortega and by modern lectin-binding methods. In response to injury, reactive microglia exhibit hypertrophy and hyperplasia, and may or may not go on to form typical lipid-laden phagocytes. Activated microglia show upregulation of the many marker antigens they share with circulating monocytes, including the major histocompatibility class (MHC) class II antigens that bespeak their immunocompetent nature. However, MHC class I and II expression and development of immunohistochemical positivity for cytoplasmic and plasma membrane antigens that characterize the monocyte-macrophage do not necessarily indicate an immunological response though there is ample evidence that microglia can serve as antigen-presenting cells. Rather, microglia are extraordinarily sensitive to changes in the brain microenvironment, whatever the nature of the exciting mechanism or substance. They may be considered to serve an ever alert, protective and supportive function that can be assembled rapidly to deal with infections, physical injuries, physiologic changes and systemic influences. In addition to elaboration and secretion of cytokines with varied actions, e.g., suppression of astrogliosis, they secrete factors, including nerve growth factor, which are supportive of neurons. They have an important role in iron metabolism and the storage of iron and ferritin. They may promote central nervous system regeneration. They are prominently involved in such pathologic processes as the acquired immunodeficiency syndrome, multiple sclerosis, prion diseases and the degenerative disorders, e.g., Alzheimer's disease and Parkinson's disease. With aging, they grow more numerous, become richer in iron and ferritin and exhibit phenotypic alteration, e.g., the expression of MHC class II antigens that are not ordinarily demonstrable immunohistochemically in the resting state. The rate of growth of our knowledge of microglia during the last decade has been exponential and continues.
Collapse
Affiliation(s)
- K D Barron
- Department of Neurology, Albany Medical College, NY 12208, USA
| |
Collapse
|
46
|
Abstract
Cytokines are a group of molecules with an extremely broad range of activities on a variety of target cells. This review summarizes the known cytokine and cytokine receptor expression in primary brain tumors and derived cell lines. These expression patterns are compared with those occurring in other CNS diseases, such as virus or bacterial infections, experimental allergic encephalitis, multiple sclerosis, and trauma. A variety of cytokines are expressed during CNS neoplasia; their potential involvement in tumor growth through a variety of mechanisms, such as autocrine or paracrine growth stimulation, angiogenesis, and immune surveillance evasion, are discussed. Finally, results of preliminary therapeutic approaches with cytokines are critically evaluated.
Collapse
Affiliation(s)
- E G Van Meir
- Neurosurgery Department, University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
47
|
Kaaijk P, Troost D, Das PK, Leenstra S, Bosch DA. Long-term culture of organotypic multicellular glioma spheroids: a good culture model for studying gliomas. Neuropathol Appl Neurobiol 1995; 21:386-91. [PMID: 8632833 DOI: 10.1111/j.1365-2990.1995.tb01075.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Gliomas, as well as other solid tumours, contain tumour stroma composed of connective tissue, macrophages, capillaries and other non-cellular constituents. Therefore, a homogeneous culture of tumour cells alone, as is often used as a culture model for gliomas, is not ideal to study all aspects of gliomas. In the present study we describe an alternative culture model, i.e. organotypic multicellular spheroids (OMS), that histologically closely resembles the tumour in vivo. Glioma explants, obtained at surgery from five patients, were cultured on agarose to form OMS, which were cultured for up to 16 weeks. At regular intervals, OMS were fixed and histological and immunocytochemical analyses were carried out. The histology as well as the immunocytochemical characteristics of the OMS proved to be almost unchanged after a culture period of 16 weeks. In contrast to monolayer cultures, glial fibrillary acidic protein (GFAP) expression in the OMS is preserved after 16 weeks of culture. However, in OMS from three out of five patients, small GFAP-negative cells appeared in the outer cell layers between 1 and 2 weeks of culture. Furthermore, after about 6 weeks of culture, the capillaries disappeared from the OMS. After prolonged culture, tumour cell heterogeneity, the cellular composition, and the histology of the OMS still closely resembled the tumour in vivo. It is suggested that OMS provide a good long-term culture model for the study of gliomas.
Collapse
Affiliation(s)
- P Kaaijk
- Department of Neurosurgery, Academic Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
48
|
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: 158] [Impact Index Per Article: 5.3] [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
|
49
|
Nakano A, Tani E, Miyazaki K, Yamamoto Y, Furuyama J. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in human gliomas. J Neurosurg 1995; 83:298-307. [PMID: 7616276 DOI: 10.3171/jns.1995.83.2.0298] [Citation(s) in RCA: 176] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The gene expression of five matrix metalloproteinases (MMPs) and two tissue inhibitors of metalloproteinases (TIMPs) was studied in human gliomas in vivo and in vitro to evaluate their roles in glioma invasion. Simultaneous expression of one to four MMP genes and two TIMP genes was found in 17 surgical glioma specimens, and one MMP (gelatinase A) gene and two TIMP genes were simultaneously expressed in tissue of three brains. The concomitant overexpression of gelatinase A, gelatinase B, and occasional matrilysin genes was associated with the malignancy of gliomas and accompanied by overexpression of the TIMP-1 gene. In five human glioma cell lines, gelatinase A, TIMP-1, and TIMP-2 genes were constitutively expressed in alll cell lines: the matrilysin gene in three cell lines; the stromelysin gene in two cell lines; and the interstitial collagenase gene in one cell line. There was a clear difference in the expression of gelatinase B and stromelysin genes between surgical glioma specimens and glioma cell lines: the gelatinase B gene was not expressed constitutively in vitro but was overexpressed in vivo, whereas the stromelysin gene was not expressed in vivo but was expressed in some cell lines. To find the cause of that difference in vivo and in vitro, the transcriptional regulations of MMP and TIMP genes by tumor promoter, growth factors, or cytokines were studied in vitro. Interstitial collagenase, gelatinase B, stromelysin, and TIMP-1 genes were upregulated in many cell lines by phorbol-12-myristate-13-acetate (PMA) and in some cell lines by epidermal growth factor, tumor necrosis factor-alpha, or interleukin-1 beta. Transforming growth factor-beta 1 (TGF beta 1) upregulated gelatinase A and matrilysin genes in some cell lines, and there were no clear responses from any MMP and TIMP genes to interleukin-6. Thus, the transcriptional modulation of MMP genes by these growth factors and cytokines seemed insufficient to explain the difference in gelatinase B and stromelysin gene expressions in vivo and in vitro and was suggestive of the genetic alteration of glioma cells in vitro, the heterogeneous cell population in glioma tissues, or both. Furthermore, the in vitro invasion of glioma cells through Matrigel in response to PMA, TGF beta 1, or TIMP-1 was assessed by chemoinvasion assay. In most cell lines, invasion was significantly stimulated by PMA or TGF beta 1 but suppressed by TIMP-1.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- A Nakano
- Department of Neurosurgery, Hyogo College of Medicine, Japan
| | | | | | | | | |
Collapse
|
50
|
Sippy BD, Hofman FM, He S, Osusky R, Sheu SJ, Walker SM, Ryan SJ, Hinton DR. SV40-immortalized and primary cultured human retinal pigment epithelial cells share similar patterns of cytokine-receptor expression and cytokine responsiveness. Curr Eye Res 1995; 14:495-503. [PMID: 7545567 DOI: 10.3109/02713689509003761] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Retinal pigment epithelial (RPE) cells produce and respond to a variety of cytokines; however, molecular and biochemical studies are restricted by the limited access to large numbers of pure cells and the variability associated with different donor sources. Despite success in establishing primary human RPE (HRPE) cell cultures, the inability to sustain consistent proliferation rates and morphology over several passages remains a concern. This problem was approached by using an immortalized line of simian virus (SV)40 transformed fetal HRPE cells (SVRPE). Cytokine production, receptor expression and responsiveness in the SVRPE cell line was analyzed to determine the usefulness of this model for studying HRPE-cytokine interactions. Using reverse transcriptase polymerase chain reaction (RT-PCR), HRPE and SVRPE cells demonstrated an identical pattern of interleukin-1 receptor (IL-1R), IL-2R (alpha sub-unit), IL-6R, interferon (IFN)-gamma R and tumor necrosis factor-alpha (TNF)R p55 expression. No amplification products for TNFR p75 or granulocyte/macrophage colony stimulating factor (GM-CSF)R were demonstrated in either population. IFN-gamma stimulation induced surface human leukocyte antigen (HLA)-DR in both SVRPE and HRPE, while TNF treatment induced surface expression of intercellular adhesion molecule (ICAM)-1 on SVRPE and upregulated ICAM from basal levels on HRPE. Both cell types showed amplification products for interleukin (IL)-1 beta, IL-6 and transforming growth factor (TGF)-beta 1 using RT-PCR. The bioassays demonstrated that both populations of unstimulated cells constitutively secrete very low levels of TGF-beta and no IL-6.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- B D Sippy
- Department of Pathology, University of Southern California School of Medicine, Los Angeles 90033, USA
| | | | | | | | | | | | | | | |
Collapse
|