1
|
Mazurek M, Rola R. The implications of nitric oxide metabolism in the treatment of glial tumors. Neurochem Int 2021; 150:105172. [PMID: 34461111 DOI: 10.1016/j.neuint.2021.105172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/03/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022]
Abstract
Glial tumors are the most common intracranial malignancies. Unfortunately, despite such a high prevalence, patients' prognosis is usually poor. It is related to the high invasiveness, tendency to relapse and the resistance of tumors to traditional methods of treatment. An important link in the aspect of these issues may be nitric oxide (NO) metabolism. It is a very complex mechanism with multidirectional effects on the neoplastic process. Depending on the concentration axis, it can both exert pro-tumor action as well as contribute to the inhibition of tumorigenesis. The latest observations show that the control of its metabolism can be very helpful in the development of new methods of treating gliomas, as well as in increasing the effectiveness of the agents currently used. The influence of nitric oxide and nitric oxide synthase (NOS) activity on glioma stem cells seem to be of particular importance. The use of specific inhibitors may allow the reduction of tumor growth and its tendency to relapse. Another important feature of GSCs is their conditioning of glioma resistance to traditional forms of treatment. Recent studies have shown that modulation of NO metabolism can suppress this effect, preventing the induction of radio and chemoresistance. Moreover, nitric oxide is involved in the regulation of a number of immune mechanisms. Adequate modulation of its metabolism may contribute to the induction of an anti-tumor response in the patients' immune system.
Collapse
Affiliation(s)
- Marek Mazurek
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University in Lublin, Poland.
| | - Radosław Rola
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University in Lublin, Poland
| |
Collapse
|
2
|
Ludwig N, Lotze MT. A treatise on endothelial biology and exosomes: homage to Theresa Maria Listowska Whiteside. HNO 2020; 68:71-79. [PMID: 31965194 DOI: 10.1007/s00106-019-00803-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exosomes are the current primary research focus of Dr. Theresa L. Whiteside. They are key mediators of intercellular communication in the head and neck, as well as other sites. Their effects in the tumor microenvironment are manifold and include suppression of immunity, promotion of angiogenesis, enabling of metastasis, as well as reprogramming of fibroblasts and mesenchymal stromal cells. The aim of this communication is to summarize Dr. Whiteside's contribution to the field of exosome research and details the interactions of exosomes with endothelial cells leading to recent findings on how to target endothelial cells using exosomes as a therapeutic approach.
Collapse
Affiliation(s)
- N Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Departments of Surgery, Cardiothoracic Surgery, Bioengineering and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - M T Lotze
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,UPMC Hillman Cancer Center, G.27A, 5150 Centre Ave, 15213, Pittsburgh, PA, USA. .,Departments of Surgery, Cardiothoracic Surgery, Bioengineering and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Chen PY, Wu CYJ, Fang JH, Chen HC, Feng LY, Huang CY, Wei KC, Fang JY, Lin CY. Functional Change of Effector Tumor-Infiltrating CCR5 +CD38 +HLA-DR +CD8 + T Cells in Glioma Microenvironment. Front Immunol 2019; 10:2395. [PMID: 31649684 PMCID: PMC6794477 DOI: 10.3389/fimmu.2019.02395] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/24/2019] [Indexed: 11/13/2022] Open
Abstract
Human glioma facilitates an impaired anti-tumor immunity response, including defects in circulation of T lymphocytes. The level of CD8+ T-cell activation acts as an immune regulator associated with disease progression. However, little is known about the characteristics of peripheral and tumor-infiltrating CD8+ T cells in patients with glioma. In this study, we examined the level of CD8+ T-cell activation in a group of 143 patients with glioma and determined that peripheral CD3+ T cells decreased in accordance with disease severity. The patients' peripheral CD8+ T-cell populations were similar to that of healthy donors, and a small amount of CD8+ tumor-infiltrating lymphocytes was identified in glioma tissues. An increase in activated CD8+ T cells, characterized as CD38+HLA-DR+, and their association with disease progression were identified in the patients' peripheral blood and glioma, and shown to display enriched CCR5+ and TNFR2+ expression levels. Ex vivo examination of CD38+HLA-DR+CD8+ T cells indicated that this subset of cells displayed stronger secretion of IFN-γ and IL-2 before and after a 6-h stimulation with phorbol 12-myristate 13-acetate (PMA) and ionomycin (ION) relative to healthy CD38+HLA-DR+CD8+ T cells, indicating the functional feasibility of CD38+HLA-DR+CD8+ T cells. Higher CCL5 protein and mRNA levels were identified in glioma tissues, which was consistent with the immunohistochemistry results revealing both CCL5 and CD38+HLA-DR+CD8+ T cell expression. Patients' CCR5+CD38+HLA-DR+CD8+ T cells were further validated and shown to display increases in CD45RA+CCR7- and T-bet+ accompanied by substantial CD107-a, IFN-γ, and Granzyme B levels in response to glioma cells.
Collapse
Affiliation(s)
- Pin-Yuan Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.,Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jian-He Fang
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsiu-Chi Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Ying Feng
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yen Lin
- School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
4
|
Madsen SJ, Christie C, Huynh K, Peng Q, Uzal FA, Krasieva TB, Hirschberg H. Limiting glioma development by photodynamic therapy-generated macrophage vaccine and allo-stimulation: an in vivo histological study in rats. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-7. [PMID: 29417766 PMCID: PMC5802332 DOI: 10.1117/1.jbo.23.2.028001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/19/2018] [Indexed: 06/08/2023]
Abstract
Immunotherapy of brain tumors involves the stimulation of an antitumor immune response. This type of therapy can be targeted specifically to tumor cells thus sparing surrounding normal brain. Due to the presence of the blood-brain barrier, the brain is relatively isolated from the systemic circulation and, as such, the initiation of significant immune responses is more limited than other types of cancers. The purpose of this study was to show that the efficacy of tumor primed antigen presenting macrophage (MaF98) vaccines can be increased by: (1) photodynamic therapy (PDT) of the priming tumor cells and (2) intracranial injection of allogeneic glioma cells directly into the tumor site. Experiments were conducted in an in vivo brain tumor development model using Fischer rats and F98 (syngeneic) and BT4C (allogeneic) glioma cells. The results showed that immunization with Ma (acting as antigen-presenting cells), primed with PDT-treated tumor cells (MaF98), significantly slowed but did not prevent the growth of F98-induced tumors in the brain. Complete suppression of tumor development was obtained via MaF98 inoculation combined with direct intracranial injection of allogeneic glioma cells. No deleterious effects were noted in any of the animals during the 14-day observation period.
Collapse
Affiliation(s)
- Steen J. Madsen
- University of Nevada, Department of Health Physics and Diagnostic Sciences, Las Vegas, Nevada, United States
| | - Catherine Christie
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Khoi Huynh
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Qian Peng
- University of Oslo, Pathology Clinic, Rikshospitalet-Radiumhospitalet HF Medical Center, Montebello, Oslo, Norway
| | - Francisco A. Uzal
- University of California, School of Veterinary Medicine, Davis, San Bernardino, California, United States
| | - Tatiana B. Krasieva
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Henry Hirschberg
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| |
Collapse
|
5
|
Schiffer D, Mellai M, Bovio E, Annovazzi L. The neuropathological basis to the functional role of microglia/macrophages in gliomas. Neurol Sci 2017; 38:1571-1577. [PMID: 28593528 DOI: 10.1007/s10072-017-3002-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022]
Abstract
The paper wants to be a tracking shot of the main recent acquisitions on the function and significance of microglia/macrophages in gliomas. The observations have been principally carried out on in vitro cultures and on tumor transplants in animals. Contrary to what is deduced from microglia in non-neoplastic pathologic conditions of central nervous system (CNS), most conclusions indicate that microglia acts favoring tumor proliferation through an immunosuppression induced by glioma cells. By immunohistochemistry, different microglia phenotypes are recognized in gliomas, from ramified microglia to frank macrophagic aspect. One wonders whether the functional conclusions drawn from many microglia studies, but not in conditions of human pathology, apply to all the phenotypes recognizable in them. It is difficult to verify in human pathology a prognostic significance of microglia. Only CD163-positive microglia/macrophages inversely correlate with glioma patients' survival, whereas the total number of microglia does not change with the malignancy grade.
Collapse
Affiliation(s)
- Davide Schiffer
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy.
| | - Marta Mellai
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| | - Enrica Bovio
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| | - Laura Annovazzi
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| |
Collapse
|
6
|
Liu Z, Meng Q, Bartek J, Poiret T, Persson O, Rane L, Rangelova E, Illies C, Peredo IH, Luo X, Rao MV, Robertson RA, Dodoo E, Maeurer M. Tumor-infiltrating lymphocytes (TILs) from patients with glioma. Oncoimmunology 2016; 6:e1252894. [PMID: 28344863 DOI: 10.1080/2162402x.2016.1252894] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 12/11/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) may represent a viable source of T cells for the biological treatment of patients with gliomas. Glioma tissue was obtained from 16 patients, tumor cell lines were established, and TILs were expanded in 16/16 cases using a combination of IL-2/IL-15/IL-21. Intracellular cytokine staining (ICS, IL-2, IL-17, TNFα and IFNγ production) as well as a cytotoxicity assay was used to detect TIL reactivity against autologous tumor cells or shared tumor-associated antigens (TAAs; i.e., NY-ESO-1, Survivin or EGFRvIII). TILs were analyzed by flow cytometry, including T-cell receptor (TCR) Vβ family composition, exhaustion/activation and T-cell differentiation markers (CD45RA/CCR7). IL-2/IL-15/IL-21 expanded TILs exhibited a mixture of CD4+, CD8+, as well as CD3+ CD4-CD8- T cells with a predominant central memory CD45RA-CCR7+ phenotype. TIL showed low frequencies of T cells testing positive for PD-1, TIM-3 and CTLA-4. LAG3 tested positive in up to 30% of CD8+ TIL, with low (1.25%) frequencies in CD4+ T cells. TIL cultures exhibited preferential usage of Vβ families and recognition of autologous tumor cells defined by cytokine production and cytotoxicity. IL-2/IL-15/IL-21 expanded TILs represent a viable source for the cellular therapy of patients with gliomas.
Collapse
Affiliation(s)
- Zhenjiang Liu
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Qingda Meng
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Jiri Bartek
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden; Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Thomas Poiret
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Oscar Persson
- Department of Neurosurgery, Karolinska University Hospital , Stockholm, Sweden
| | - Lalit Rane
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Elena Rangelova
- Department of Clinical Science, Intervention and Technology, Karolinska Hospital , Stockholm, Sweden
| | - Christopher Illies
- Department of Neurosurgery, Karolinska University Hospital , Stockholm, Sweden
| | - Inti Harvey Peredo
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden
| | - Xiaohua Luo
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Martin Vijayakumar Rao
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Rebecca Axelsson Robertson
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden
| | - Ernest Dodoo
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden
| | - Markus Maeurer
- Therapeutic Immunology Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
7
|
Ding P, Wang W, Wang J, Yang Z, Xue L. Expression of tumor-associated macrophage in progression of human glioma. Cell Biochem Biophys 2015; 70:1625-31. [PMID: 25182001 DOI: 10.1007/s12013-014-0105-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aim of this study is to investigate the expression of tumor-associated macrophages (TAMs) M1, M2 phenotypic in human glioma tissues, and to explore the clinical significance and prognostic value of TAMs in glioma patients. A total of 50 glioma samples were obtained from patients diagnosed in our hospital from 2007 to 2010. Clinical follow-up was conducted via return visits and telephone interviews after discharge. Progression free survival (PFS) was calculated based on tumor progression by MRI and CT examination from the primary operation. Overall survival (OS) time was calculated from the initial surgical operation date to end date of follow-up or death. Kaplan-Meier methodology was used to evaluate the survival of patients and log-rank test for comparing differences between groups. The expression levels of CD16 and CD206 were investigated in the 4 μm serial paraffin sections by immunohistochemistry. M1-type macrophages filtrated in all the grades of glioma samples, and the lower expression level was associated with high grade glioma. A negative correlation was found between WHO pathological grades and the expression of M1-type macrophages by Spearman correlation analysis. M2-type macrophages filtrated in all the grades of glioma samples with the higher expression level associated with high grade glioma. A positive correlation was found between WHO pathological grades and the expression of M2-type macrophages by Spearman correlation analysis. The PFS and OS among patients with high levels of M1-type macrophages (CD16+++) were significantly higher than those with less expression. The PFS and OS among patients with high levels of M2-type macrophages (CD206+++) were significantly lower than those with low expression. M1-type macrophages may inhibit the tumor growth and improve the therapeutic outcome of glioma patients. M2 ratios are associated with tumor proliferation and poor prognosis. TAMs phenotypes of glioma samples are the potential biomarkers in assessing the degree of malignancy, tumor invasion, and patient prognosis in clinic.
Collapse
Affiliation(s)
- Peng Ding
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, 295 XiChang Rd, Kunming, 650032, People's Republic of China
| | | | | | | | | |
Collapse
|
8
|
Paff M, Alexandru-Abrams D, Hsu FPK, Bota DA. The evolution of the EGFRvIII (rindopepimut) immunotherapy for glioblastoma multiforme patients. Hum Vaccin Immunother 2014; 10:3322-31. [PMID: 25625931 PMCID: PMC4514075 DOI: 10.4161/21645515.2014.983002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/08/2014] [Accepted: 10/19/2014] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is the most common type of brain tumor and it is uniformly fatal. The community standard of treatment for this disease is gross or subtotal resection of the tumor, followed by radiation and temozolomide. At recurrence bevacizumab can be added for increased progression free survival. Many challenges are encountered while trying to devise new drugs to treat GBM, such as the presence of the blood brain barrier which is impermeable to most drugs. Therefore in the past few years attention was turned to immunological means for the treatment of this devastating disease. EGFRvIII targeting has proven a good way to attack glioblastoma cells by using the immune system. Although in still in development, this approach holds the promise as a great first step toward immune-tailored drugs for the treatment of brain cancers.
Collapse
Key Words
- ACTIVATE, A Complementary Trial of an Immunotherapy Vaccine against Tumor Specific EGRFvIII
- APC, antigen-presenting cell
- Ab, antibody
- BBB, blood brain barrier
- CD25, cluster of differentiation 25
- CD4, cluster of differentiation 4
- CNS, central nervous system
- CPT-11, irinotecan, Camptosar
- CTL, Cytotoxic T lymphocytes
- D, day
- DTH, delayed-type hypersensitivity
- EGFRVIII
- EGFRvIII, The epidermal growth factor receptor variant III
- EORTC, European Organization for Research and Treatment of Cancer
- GAGE, G antigen gene family
- GBM, Glioblastoma Multiforme
- GM-CSF, Granulocyte-macrophage colony-stimulating factor
- Grb2, Growth factor receptor-bound protein 2
- HLA, human leukocyte antigen
- IL-10, Interleukin-10
- IL-12, Interleukin-12
- IL-2, Interleukin-2
- INF-g, Interferon gamma
- KLH, keyhole limpet hemocyanin
- KPS, Karnofsky performance status
- LPS, lipopolysaccharide
- MGMT, O-6-methylguanine-DNA methyltransferase
- MHC, major histocompatibility complex
- NCIC, National Cancer Institute of Canada
- OS, overall survival
- PFS, progression-free survival
- PGE2, prostaglandin E2
- Ras, rat sarcoma genes
- SEER, Surveillance, Epidemiology, and End Results Program
- TGF-b, transforming growth factor beta
- TH2 cells, T helper type 2 cells
- TMZ, temozolomide
- TTP, time to progression
- Treg cells, regulatory T cells
- VEGF, Vascular endothelial growth factor
- WHO, World Health Organization
- Y, year
- brain
- glioblastoma
- therapies
- vaccine
Collapse
Affiliation(s)
- Michelle Paff
- Department of Neurological Surgery; University of California, Irvine; Orange, CA USA
| | | | - Frank P K Hsu
- Department of Neurological Surgery; University of California, Irvine; Orange, CA USA
| | - Daniela A Bota
- Department of Neurological Surgery; University of California, Irvine; Orange, CA USA
- Department of Neurology; University of California, Irvine; Orange, CA USA
- Chao Family Comprehensive Cancer Center; University of California, Irvine; Orange, CA USA
| |
Collapse
|
9
|
Bielamowicz K, Khawja S, Ahmed N. Adoptive cell therapies for glioblastoma. Front Oncol 2013; 3:275. [PMID: 24273748 PMCID: PMC3823029 DOI: 10.3389/fonc.2013.00275] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/25/2013] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is the most common and most aggressive primary brain malignancy and, as it stands, is virtually incurable. With the current standard of care, maximum feasible surgical resection followed by radical radiotherapy and adjuvant temozolomide, survival rates are at a median of 14.6 months from diagnosis in molecularly unselected patients (1). Collectively, the current knowledge suggests that the continued tumor growth and survival is in part due to failure to mount an effective immune response. While this tolerance is subtended by the tumor being utterly “self,” it is to a great extent due to local and systemic immune compromise mediated by the tumor. Different cell modalities including lymphokine-activated killer cells, natural killer cells, cytotoxic T lymphocytes, and transgenic chimeric antigen receptor or αβ T cell receptor grafted T cells are being explored to recover and or redirect the specificity of the cellular arm of the immune system toward the tumor complex. Promising phase I/II trials of such modalities have shown early indications of potential efficacy while maintaining a favorable toxicity profile. Efficacy will need to be formally tested in phase II/III clinical trials. Given the high morbidity and mortality of GBM, it is imperative to further investigate and possibly integrate such novel cell-based therapies into the current standards-of-care and herein we collectively assess and critique the state-of-the-knowledge pertaining to these efforts.
Collapse
Affiliation(s)
- Kevin Bielamowicz
- Center for Cell and Gene Therapy, Baylor College of Medicine , Houston, TX , USA ; Texas Children's Cancer Center, Baylor College of Medicine , Houston, TX , USA ; Department of Pediatrics, Baylor College of Medicine , Houston, TX , USA ; Baylor College of Medicine , Houston, TX , USA
| | | | | |
Collapse
|
10
|
Tumor-associated macrophages in glioma: friend or foe? JOURNAL OF ONCOLOGY 2013; 2013:486912. [PMID: 23737783 PMCID: PMC3664503 DOI: 10.1155/2013/486912] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/08/2013] [Indexed: 12/31/2022]
Abstract
Tumor-associated macrophages (TAMs) contribute substantially to the tumor mass of gliomas and have been shown to play a major role in the creation of a tumor microenvironment that promotes tumor progression. Shortcomings of attempts at antiglioma immunotherapy may result from a failure to adequately address these effects. Emerging evidence supports an independent categorization of glioma TAMs as alternatively activated M2-type macrophages, in contrast to classically activated proinflammatory M1-type macrophages. These M2-type macrophages exert glioma-supportive effects through reduced anti-tumor functions, increased expression of immunosuppressive mediators, and nonimmune tumor promotion through expression of trophic and invasion-facilitating substances. Much of our work has demonstrated these features of glioma TAMs, and together with the supporting literature will be reviewed here. Additionally, the dynamics of glioma cell-TAM interaction over the course of tumor development remain poorly understood; our efforts to elucidate glioma cell-TAM dynamics are summarized. Finally, the molecular pathways which underlie M2-type TAM polarization and gene expression similarly require further investigation, and may present the most potent targets for immunotherapeutic intervention. Highlighting recent evidence implicating the transcription factor STAT3 in immunosuppressive tumorigenic glioma TAMs, we advocate for gene array-based approaches to identify yet unappreciated expression regulators and effector molecules important to M2-type glioma TAMs polarization and function within the glioma tumor microenvironment.
Collapse
|
11
|
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
|
12
|
|
13
|
Jotereau F, Gervois N, Labarrière N. Adoptive transfer with high-affinity TCR to treat human solid tumors: how to improve the feasibility? Target Oncol 2012; 7:3-14. [PMID: 22350487 DOI: 10.1007/s11523-012-0207-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 01/12/2012] [Indexed: 01/05/2023]
Abstract
The adoptive transfer of tumor antigen-specific T cells recently achieved clinical efficacy for a fraction of melanoma patients refractory to other therapies. Unfortunately, the application of this strategy to the remaining melanoma and most other cancer patients is hampered by the difficulty to generate high-affinity tumor-reactive T cells. Two strategies are currently developed to extend the feasibility of this therapeutic approach: clinical grade tool production for MHC-peptide multimer-driven sorting of antigen-specific T cells from the endogenous peripheral T cell repertoire and de novo engineering of the missing repertoire by genetic transfer of cloned specific T cell receptor (TCR) into T cells. The expected multiplication of adoptive transfer treatments, by these strategies, and their careful evaluation should enable the cure of a number of otherwise compromised cancer patients and to gain insight into the characteristics of transferred T cells best fitted to eradicate tumor cells, in terms of antigen specificities, phenotype, and functions. In particular, identification of tumor-rejection antigens by this approach would improve the design and efficacy of all immunotherapeutic approaches.
Collapse
|
14
|
Marshall D, Mitchell DA, Graner MW, Bigner DD. Immunotherapy of brain tumors. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:309-30. [PMID: 22230450 DOI: 10.1016/b978-0-444-52138-5.00020-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
15
|
Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, Henry V, Colman H, Priebe W, Sawaya R, Lang FF, Heimberger AB. Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway. Mol Cancer Ther 2010; 9:67-78. [PMID: 20053772 DOI: 10.1158/1535-7163.mct-09-0734] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma multiforme (GBM) is a lethal cancer that responds poorly to radiotherapy and chemotherapy. Glioma cancer-initiating cells have been shown to recapitulate the characteristic features of GBM and mediate chemotherapy and radiation resistance. However, it is unknown whether the cancer-initiating cells contribute to the profound immune suppression in GBM patients. Recent studies have found that the activated form of signal transducer and activator of transcription 3 (STAT3) is a key mediator in GBM immunosuppression. We isolated and generated CD133+ cancer-initiating single colonies from GBM patients and investigated their immune-suppressive properties. We found that the cancer-initiating cells inhibited T-cell proliferation and activation, induced regulatory T cells, and triggered T-cell apoptosis. The STAT3 pathway is constitutively active in these clones and the immunosuppressive properties were markedly diminished when the STAT3 pathway was blocked in the cancer-initiating cells. These findings indicate that cancer-initiating cells contribute to the immune evasion of GBM and that blockade of the STAT3 pathway has therapeutic potential.
Collapse
Affiliation(s)
- Jun Wei
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77230-1402, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Han SJ, Kaur G, Yang I, Lim M. Biologic Principles of Immunotherapy for Malignant Gliomas. Neurosurg Clin N Am 2010; 21:1-16. [DOI: 10.1016/j.nec.2009.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
17
|
The role of tregs in glioma-mediated immunosuppression: potential target for intervention. Neurosurg Clin N Am 2009; 21:125-37. [PMID: 19944972 DOI: 10.1016/j.nec.2009.08.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of regulatory T cells (Tregs) in mediating immune suppression of anti-tumor immune responses is increasingly appreciated in patients with malignancies-especially within the malignant glioma patient population. This article discuss the role and prognostic significance of Tregs within glioma patients and delineates potential approaches for their inhibition that can be used alone or in combination with other immune therapeutics in clinical trials and in the clinical settings of recurrent or residual disease.
Collapse
|
18
|
Abstract
Conventional therapies for glioblastoma multiforme (GBM) fail to target tumor cells exclusively, resulting in non-specific toxicity. Immune targeting of tumor-specific mutations may allow for more precise eradication of neoplastic cells. EGFR variant III (EGFRvIII) is a tumor-specific mutation that is widely expressed in GBM and other neoplasms and its expression enhances tumorigenicity. This in-frame deletion mutation splits a codon, resulting in a novel glycine at the fusion junction producing a tumor-specific epitope target for cellular or humoral immunotherapy. We have previously shown that vaccination with a peptide that spans the EGFRvIII fusion junction (PEPvIII-KLH/CDX-110) is an efficacious immunotherapy in syngeneic murine models. In this review, we summarize our results in GBM patients targeting this mutation in multiple, multi-institutional Phase II immunotherapy trials. These trials demonstrated that a selected population of GBM patients who received vaccines targeting EGFRvIII had an unexpectedly long survival time. Further therapeutic strategies and potential pitfalls of using this approach are discussed.
Collapse
Affiliation(s)
- Amy B Heimberger
- University of Texas MD Anderson Cancer Center, Department of Neurosurgery, Unit 422, Houston, TX 77230-1402, USA.
| | | |
Collapse
|
19
|
Sampson JH, Archer GE, Mitchell DA, Heimberger AB, Bigner DD. Tumor-specific immunotherapy targeting the EGFRvIII mutation in patients with malignant glioma. Semin Immunol 2008; 20:267-75. [PMID: 18539480 PMCID: PMC2633865 DOI: 10.1016/j.smim.2008.04.001] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 04/08/2008] [Accepted: 04/17/2008] [Indexed: 01/07/2023]
Abstract
Conventional therapies for malignant gliomas (MGs) fail to target tumor cells exclusively, such that their efficacy is ultimately limited by non-specific toxicity. Immunologic targeting of tumor-specific gene mutations, however, may allow more precise eradication of neoplastic cells. The epidermal growth factor receptor variant III (EGFRvIII) is a consistent tumor-specific mutation that is widely expressed in MGs and other neoplasms. This mutation encodes a constitutively active tyrosine kinase that enhances tumorgenicity and migration and confers radiation and chemotherapeutic resistance. This in-frame deletion mutation splits a codon resulting in the creation of a novel glycine at the fusion junction between normally distant parts of the molecule and producing a sequence re-arrangement which creates a tumor-specific epitope for cellular or humoral immunotherapy in patients with MGs. We have previously shown that vaccination with a peptide that spans the EGFRvIII fusion junction is an efficacious immunotherapy in syngeneic murine models, but patients with MGs have a profound immunosuppression that may inhibit the ability of antigen presenting cells (APCs), even those generated ex vivo, to induce EGFRvIII-specific immune responses. In this report, we summarize our results in humans targeting this mutation in two consecutive and one multi-institutional Phase II immunotherapy trials. These trials demonstrated that vaccines targeting EGFRvIII are capable of inducing potent T- and B-cell immunity in these patients, and lead to an unexpectedly long survival time. Most importantly, vaccines targeting EGFRvIII were universally successful at eliminating tumor cells expressing the targeted antigen without any evidence of symptomatic collateral toxicity. These studies establish the tumor-specific EGFRvIII mutation as a novel target for humoral- and cell-mediated immunotherapy in a variety of cancers. The recurrence of EGFRvIII-negative tumors in our patients, however, highlights the need for targeting a broader repertoire of tumor-specific antigens.
Collapse
Affiliation(s)
- John H Sampson
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | |
Collapse
|
20
|
Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, Yang DS, Sun W, Qiao W, Hiraoka N, Fuller GN. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res 2008; 14:5166-72. [PMID: 18698034 DOI: 10.1158/1078-0432.ccr-08-0320] [Citation(s) in RCA: 258] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The incidence of regulatory T cells (Treg) in intrinsic central nervous system malignancies is unknown. Immunotherapeutic approaches that inhibit the Treg population may be limited to a subset of patients with gliomas. Our hypothesis is that only the most malignant gliomas have a prominent glioma-infiltrating Treg population that contributes to the immunosuppressive biology and that the presence of Tregs is a negative prognostic variable. EXPERIMENTAL DESIGN We measured the incidence of Tregs in 135 glial tumors (including all pathologic types) in a glioma microarray using immunohistochemical analysis. Results were categorized according to the total number of Tregs within the tumors. Correlation of the presence of Tregs with prognosis was evaluated using univariate and multivariate analyses. RESULTS Tregs were not present in normal brain tissue and were very rarely found in low-grade gliomas and oligodendrogliomas. We observed significant differences in the prevalence of Tregs between astrocytic and oligodendroglial tumors, between tumors of different grades, and between different pathologic types of tumors. We identified Tregs most frequently in glioblastoma multiforme (GBM) but very rarely in low-grade astrocytomas. The presence of Tregs within GBMs did not alter the median survival in patients from whom the tumors were obtained. CONCLUSIONS Treg infiltration differed significantly in the tumors according to lineage, pathology, and grade. Tregs seemed to have the highest predilection for tumors of the astrocytic lineage and specifically in the high-grade gliomas, such as GBM. In both univariate and multivariate analysis, the presence of Tregs in GBMs seemed to be prognostically neutral.
Collapse
Affiliation(s)
- Amy B Heimberger
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Despite aggressive multi-modality therapy including surgery, radiation, and chemotherapy, the prognosis for patients with malignant primary brain tumors remains very poor. Moreover, the non-specific nature of conventional therapy for brain tumors often results in incapacitating damage to surrounding normal brain and systemic tissues. Thus, there is an urgent need for the development of therapeutic strategies that precisely target tumor cells while minimizing collateral damage to neighboring eloquent cerebral cortex. The rationale for using the immune system to target brain tumors is based on the premise that the inherent specificity of immunologic reactivity could meet the clear need for more specific and precise therapy. The success of this modality is dependent on our ability to understand the mechanisms of immune regulation within the central nervous system (CNS), as well as counter the broad defects in host cell-mediated immunity that malignant gliomas are known to elicit. Recent advances in our understanding of tumor-induced and host-mediated immunosuppressive mechanisms, the development of effective strategies to combat these suppressive effects, and a better understanding of how to deliver immunologic effector molecules more efficiently to CNS tumors have all facilitated significant progress toward the realization of true clinical benefit from immunotherapeutic treatment of malignant gliomas.
Collapse
Affiliation(s)
- Duane A Mitchell
- Division of Neurosurgery, Department of Surgery, The Preston Robert Tisch Brain Tumor Center, Duke, NC 27710, USA.
| | | | | |
Collapse
|
22
|
Li B, Lin J, Vanroey M, Jure-Kunkel M, Jooss K. Established B16 tumors are rejected following treatment with GM-CSF-secreting tumor cell immunotherapy in combination with anti-4-1BB mAb. Clin Immunol 2007; 125:76-87. [PMID: 17706463 DOI: 10.1016/j.clim.2007.07.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 06/07/2007] [Accepted: 07/09/2007] [Indexed: 01/30/2023]
Abstract
Immunization with irradiated tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulates potent, specific and long lasting anti-tumor immunity in clinical and preclinical settings. Efforts to further increase immunotherapy efficacy with immune-modulatory agents are under evaluation. Based on the immune-modulatory properties of 4-1BB (CD137), it has been postulated that agonistic 4-1BB antibodies may add additional anti-tumor efficacy to GM-CSF-secreting tumor cell immunotherapy. The combination of GM-CSF-secreting tumor cell immunotherapy and anti-4-1BB monoclonal antibody (mAb) treatment resulted in rejection of established tumors in the B16 melanoma model. These anti-tumor effects correlated with persistent tumor-specific CD8(+) T cell responses. In addition, early tumor infiltration of functional CD8(+) T cells and a greater expansion of antigen-specific memory T cells were found in mice treated with the combination therapy. In summary, an agonistic anti-4-1BB mAb combined with GM-CSF-secreting tumor cell immunotherapy may provide a novel and potent treatment strategy for patients with cancer.
Collapse
Affiliation(s)
- Betty Li
- Cell Genesys Inc, South San Francisco, CA 94080, USA.
| | | | | | | | | |
Collapse
|
23
|
Liu Y, Wang Q, Kleinschmidt-DeMasters BK, Franzusoff A, Ng KY, Lillehei KO. TGF-beta2 inhibition augments the effect of tumor vaccine and improves the survival of animals with pre-established brain tumors. J Neurooncol 2006; 81:149-62. [PMID: 16941073 DOI: 10.1007/s11060-006-9222-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Accepted: 07/17/2006] [Indexed: 10/24/2022]
Abstract
TGF-beta2 secretion by high grade gliomas has been implicated as one of the major factors contributing to tumor growth, alterations in the host immune response to tumor, and failure of gliomas to respond to current immunotherapy strategies. We hypothesized that targeted delivery and inhibition of TGF-beta2 by TGF-beta2 antisense oligonucleotides (AS-ODNs) would overcome tumor-induced immunosuppression and enhance the capacity of tumor vaccines to eradicate established brain tumors. Utilizing the mRNA sequences of TGF-beta2, specific AS-ODNs were constructed and tested for their ability to inhibit TGF-beta2 production in 9L glioma cells. The effect of combining local intracranial administration of antisense ODNs with systemic tumor vaccine was examined. Fisher 344 rats were vaccinated subcutaneously with irradiated 9L tumor cells 3 days after intracranial tumor implantation. Four days after vaccination, ODNs were administered into the tumor mass and survival was followed. ODNs delivered locally distributed widely within the brain tumor mass and inhibited TGF-beta2 expression. Survival of tumor-bearing rats treated with the combination of local antisense and systemic tumor vaccine was significantly enhanced (mean survival time (MST): 48.0 days). In contrast, MST for animals treated with nonsense plus vaccine, vaccine alone, antisense alone or PBS showed no survival advantage and no statistical differences between groups (33.5 days, 29.0 days, 37.5 days, and 31.5 days, respectively). Our data supports the hypothesis that local administration of antisense TGF-beta2 ODNs combined with systemic vaccination can increase efficacy of immunotherapy and is a novel, potentially clinically applicable, strategy for high-grade glioma treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurosurgery, C-307, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Denver, CO, 80262, USA
| | | | | | | | | | | |
Collapse
|
24
|
Fakhrai H, Mantil JC, Liu L, Nicholson GL, Murphy-Satter CS, Ruppert J, Shawler DL. Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther 2006; 13:1052-60. [PMID: 16826191 DOI: 10.1038/sj.cgt.7700975] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We performed a phase I clinical trial in grade IV astrocytoma to assess the safety of a whole-cell vaccine comprising autologous tumor cells genetically modified by a transforming growth factor-beta2 (TGF-beta2) antisense vector. Blocking secretion of the immunosuppressive molecule TGF-beta in this manner should inhibit one of the major mechanisms by which tumor cells evade immune surveillance and should lead to clinically effective antitumor immunity. Six patients with progressive WHO grade IV astrocytoma were enrolled in the trial. Patients received 2-7 subcutaneous injections of 5 x 10(6)-2 x 10(7) autologous tumor cells per injection. TGF-beta2 secretion by the tumor cells used to vaccinate patients was inhibited by 53-98%. Treatment was well tolerated with only low-grade, transient treatment-related toxicities reported. Two patients had partial regressions and two had stable disease following therapy. The overall median survival was 68 weeks. Median survival of the responding patients was 78 weeks, compared to a historic value of 47 weeks for glioma patients treated conventionally. There were indications of humoral and cellular immunity induced by the vaccine. These findings support further clinical evaluation of vaccines comprised of TGF-beta antisense-modified tumor cells.
Collapse
Affiliation(s)
- H Fakhrai
- Advanced Biotherapies, Inc., San Diego, CA, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Avninder S, Sharma MC, Deb P, Mehta VS, Karak AK, Mahapatra AK, Sarkar C. Gemistocytic astrocytomas: histomorphology, proliferative potential and genetic alterations – a study of 32 cases. J Neurooncol 2006; 78:123-7. [PMID: 16614946 DOI: 10.1007/s11060-005-9077-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Accepted: 11/21/2005] [Indexed: 10/24/2022]
Abstract
Gemistocytic astrocytomas (GAs) are a distinct variant of astrocytomas, generally classified as WHO grade II, and are associated with an aggressive biological behavior. This study was undertaken to determine the histomorphological spectrum, and correlate these with their proliferative potential and genetic alterations, in order to establish a biological basis for their unfavorable prognosis.A total of 32 GAs diagnosed during an 11-year period (1993-2003) were included in the study. Immunoreactivity for CD3 (T-cells), CD20 (B-cells) and CD68 (macrophages) were evaluated to characterize the perivascular inflammatory infiltrates, while p53, epidermal growth factor receptor (EGFR), cyclin D1 and p27-immunolabeling were studied to analyze the tumor biology.Overall, the mean gemistocytic index in the study was 39.6% (range, 12.2-80.8%), with multinucleation in gemistocytes and mitosis being present in 56.2% and 15.6% respectively. Perivascular mononuclear cell cuffing was seen in 56.2% cases, which was immunopositive for CD3 and CD68 in 14 cases each, with 13 cases being immunopositive for both. Similar type of inflammatory infiltrates was also present within the tumor parenchyma. Proliferation index depicted by MIB-1 LI was low (mean: 3.7%; range: 0.5-10.5%), with 70% cases having LI of <5%. MIB-1 labeling was restricted to the small astrocytic cells, similar to p27 and cyclin D1 immunoreactivity, both of which were present in 71.5% cases. In contrast, p53 protein expression was present in 75% cases, and was strongly positive in both gemistocytes and small cells, denoting neoplastic population. However, EGFR protein expression was consistently negative in all cases. Gemistocytes lack proliferative activity possibly indicating terminal differentiation, while small cells are the proliferating cells and their overall percentage may reflect the biological aggressiveness of these tumors and help to identify GAs of higher grade undergoing malignant progression. Therefore it appears that GAs should not be uniformly graded as grade II but should be subdivided into grades II and III neoplasms based on histological features and MIB-1 LI. The poor prognosis in GAs could be attributed both to the high frequency of p53 mutations and low p27 LI.
Collapse
Affiliation(s)
- Singh Avninder
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, 110029, New Delhi, India
| | | | | | | | | | | | | |
Collapse
|
26
|
Li S, Xu J, Makarenkova VP, Tjandrawan T, Vakkila J, Reichert T, Gooding W, Lagenaur CF, Achim CL, Chambers WH, Herberman RB, Whiteside TL, Vujanovic NL. A novel epitope of N-CAM defines precursors of human adherent NK cells. J Leukoc Biol 2004; 76:1187-99. [PMID: 15356097 DOI: 10.1189/jlb.0802386] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Activated, adherent natural killer (A-NK) cells represent a distinct subpopulation of interleukin (IL)-2-stimulated NK cells, which are selectively endowed with the increased expression of integrins and ability to adhere to solid surfaces, migrate into, infiltrate, and destroy cancerous tissues. The present study defines the phenotype and functions of precursors of A-NK (pre-A-NK) cells in humans. Peripheral blood pre-A-NK cells, in contrast to the rest of NK cells, express a novel epitope of CD56 neuronal cell adhesion molecule, termed ANK-1, and increased cell-surface levels of integrins. Pre-A-NK cells also express low levels of CD56 and CD161, and some express CD162 receptor, do not express CD25 or activation markers, and are effective mediators of NK cytotoxicity. Thus, pre-A-NK cells are generally similar to CD56(dim) NK cells. However, pre-A-NK cells differ from the main NK cell subpopulation by having a lower expression level of CD16 and a lower ability to mediate redirected antibody-dependent, cell-mediated cytotoxicity. More importantly, pre-A-NK cells are preferentially endowed with the ability to rapidly respond to IL-2 by integrin-mediated adherence to endothelial cells, extracellular matrix, and plastic. This early, specific response of pre-A-NK cells to IL-2 is followed by their activation, vigorous proliferation, and differentiation into phenotypically and functionally similar A-NK cells. Pre-A-NK cells represent only approximately 26% of peripheral blood NK cells but encompass the majority of NK cells in normal and cancerous, solid tissues. We conclude that pre-A-NK cells represent a distinct subset of resting, mature NK cells with the characteristics indicative of their ability to migrate and reside in solid tissues.
Collapse
MESH Headings
- Antibodies, Monoclonal/immunology
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- CD56 Antigen/immunology
- CD56 Antigen/metabolism
- Cell Adhesion/drug effects
- Cell Adhesion/immunology
- Cell Count
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Cell Lineage/drug effects
- Cell Lineage/immunology
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cell Proliferation/drug effects
- Cells, Cultured
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/immunology
- Cytotoxicity, Immunologic/immunology
- Epitopes/immunology
- Humans
- Immunophenotyping
- Integrins/immunology
- Integrins/metabolism
- Interleukin-2/immunology
- Interleukin-2/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Lymphoid Tissue/immunology
- Lymphoid Tissue/metabolism
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- NK Cell Lectin-Like Receptor Subfamily B
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Stem Cells/drug effects
- Stem Cells/immunology
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Shen Li
- Department of Pathology, University of Pittsburgh School of Medicine, PA 15213-1863, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Despite remarkable advancements in imaging modalities and treatment options available to patients diagnosed with malignant brain tumors, the prognosis for those with high-grade lesions remains poor. The imprecise mechanisms of currently available treatments to manage these tumors do not spare damage to the normal surrounding brain and often result in major cognitive and motor deficits. Immunotherapy holds the promise of offering a potent, yet targeted, treatment to patients with brain tumors, with the potential to eradicate the malignant tumor cells without damaging normal tissues. The T cells of the immune system are uniquely capable of recognizing the altered protein expression patterns within tumor cells and mediating their destruction through a variety of effector mechanisms. Adoptive T-cell therapy is an attempt to harness and amplify the tumor-eradicating capacity of a patients' own T cells and then return these effectors to the patient in such a state that they effectively eliminate residual tumor. Although this approach is not new to the field of tumor immunology, new advancements in our understanding of T-cell activation and function and breakthroughs in tumor antigen discovery hold great promise for the translation of this modality into a clinical success.
Collapse
Affiliation(s)
- Duane A Mitchell
- Department of Pathology and Department of Surgery, Duke University Medical Center, Box 3807, Durham, NC 27710, USA
| | | | | |
Collapse
|
28
|
Yang L, Ng KY, Lillehei KO. Cell-mediated immunotherapy: a new approach to the treatment of malignant glioma. Cancer Control 2003; 10:138-47. [PMID: 12712008 DOI: 10.1177/107327480301000205] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The dismal prognosis for patients harboring intracranial gliomas has prompted an intensive search for effective treatment alternatives such as immunotherapy. Our increased knowledge in basic immunology, glioma immunobiology, and molecular biology may lead to the development of effective, rational immunotherapy approaches. METHODS The authors reviewed the literature on glioma immunology, the status of tumor vaccine therapy and on novel techniques to monitor the tumor-specific immune response. RESULTS Experimental conditions currently exist whereby potent antitumor cell-mediated immune responses can be generated. However, clinically, no therapeutic regimen has proven effective. Obstacles to establishing an effective immunotherapy regimen are the lack of a well-defined glioma-specific antigen, the heterogeneity of tumor cells in gliomas, and the modulating effect of the glioma itself on the immune system. Unique strategies to overcome these barriers are being developed. CONCLUSIONS Novel strategies to generate an anti-glioma immune response through use of dendritic cell vaccination, directed cytokine delivery, gene-based immunotherapy, and reversal of tumor-induced immunosuppression are promising. These strategies carry the potential of overcoming the resistance of gliomas to immunotherapeutic manipulation and, undoubtedly, will become a part of our future therapeutic armamentarium.
Collapse
Affiliation(s)
- Liu Yang
- Department of Neurosurgery, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | |
Collapse
|
29
|
Kito T, Kuroda E, Yokota A, Yamashita U. Cytotoxicity in glioma cells due to interleukin-12 and interleukin-18-stimulated macrophages mediated by interferon-gamma-regulated nitric oxide. J Neurosurg 2003; 98:385-92. [PMID: 12593627 DOI: 10.3171/jns.2003.98.2.0385] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Interleukin (IL)-12 and IL-18 synergistically mediate antitumor responses through the production of interferon-gamma (IFNgamma) by T and natural killer (NK) cells. Recently, it has been reported that macrophages stimulated with these cytokines also produce IFNgamma, which led the authors to investigate the antiglioma activity of macrophages stimulated by the combination of these cytokines in vitro. METHODS Dish-adherent peritoneal exudate cells, which had been elicited in thioglycollate broth as a source of macrophages, were used in the experiment. The murine glioma cell lines VM-glioma and 203G were labeled with [3H]thymidine for a cytotoxicity assay of macrophages. In response to the combined stimulation by IL-12 and IL-18, macrophages expressed potent cytotoxic activity against glioma cells in association with increasing production of IFNgamma and nitric oxide (NO). Inhibitors of NO abrogated the cytotoxic activity of the macrophages, which had been induced by IL-12 and IL-18, despite the increase in IFNgamma production. Neutralization of IFNgamma or use of macrophages obtained from IFNgamma gene-knockout mice markedly reduced not only cytotoxic activity, but also NO production. Depletion of T and NK cells from the macrophage population, which was achieved using antibody plus complement treatment, slightly reduced macrophage activities, suggesting that these are the main effector cells, although T and NK cells may partially participate in this cytotoxicity. CONCLUSIONS Macrophages stimulated with IL-12 and IL-18 produced IFNgamma and NO, which in turn mediated the antiglioma response. Therefore, macrophages as well as T and NK cells play an important role in antitumor responses stimulated by IL-12 and IL-18.
Collapse
Affiliation(s)
- Tomohiro Kito
- Departments of Immunology and Neurosurgery, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | | | | | | |
Collapse
|
30
|
Abstract
As an immunization platform for brain tumors, dendritic cells supply an impressive host of advantages. On the simplest level, they provide the safety and tumor-specificity so wanted by current therapeutic options. Yet, in addition, as the fundamental antigen-presenting cell, they circumvent many of the immunologic challenges that gliomas and the CNS proffer and that other immunotherapeutic modes fail to overcome. Directions to take now include the identification of new tumor-specific and tumor-associated antigens; the determination of the optimal dendritic cell subtype, generation, loading method, maturation state, dose, and route of delivery for immunizations; the further characterization of dendritic cells and their activities; and, potentially, the discovery of ways to pulse dendritic cells efficiently in vivo. Preclinical studies continue to play an important role in refining this form of active immunotherapy.
Collapse
Affiliation(s)
- Peter E Fecci
- Departments of Neurosurgery and Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
31
|
Loh JK, Hwang SL, Lieu AS, Huang TY, Howng SL. The alteration of prostaglandin E2 levels in patients with brain tumors before and after tumor removal. J Neurooncol 2002; 57:147-50. [PMID: 12125976 DOI: 10.1023/a:1015782809966] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Both experimental and human tumors often synthesize high levels of prostaglandins, most notably prostaglandin E2 (PGE2). This compound may play an important role in tumor growth and immunosuppression. Little is known of the production of PGE2 by brain tumors. The present study was designed to investigate the levels of PGE2 in the plasma of human brain tumors before and after tumor removal. METHODS The plasma PGE2 levels of brain tumors before and after tumor removal were measured by high-performance liquid chromatography (HPLC). RESULTS There is a significantly high concentration of PGE2 in malignant brain tumor before tumor removal. Significantly decrease of PGE2 concentration after total removal of the tumor was found both in the malignant and benign brain tumor groups (P = 0.0001 and P = 0.0039 respectively). However, compared to the control group, only malignant brain tumor showed a significant decrease of PGE2 concentration after tumor removal (P = 0.0009). CONCLUSION Our study demonstrates the malignant brain tumor synthesized higher relative proportions of PGE2 and surgical removal of the brain tumor can reduce the production of PGE2.
Collapse
Affiliation(s)
- Joon-Khim Loh
- Department of Surgery, Kaohsiung Medical University Hospital, Taiwan, ROC
| | | | | | | | | |
Collapse
|
32
|
Sanchez R, Williams C, Daza JL, Dan Q, Xu Q, Chen Y, Delgado C, Arpajirakul N, Jeffes EWB, Kim RC, Douglass T, Al Atar U, Terry Wepsic H, Jadus MR. T9 glioma cells expressing membrane-macrophage colony stimulating factor produce CD4+ T cell-associated protective immunity against T9 intracranial gliomas and systemic immunity against different syngeneic gliomas. Cell Immunol 2002; 215:1-11. [PMID: 12142031 DOI: 10.1016/s0008-8749(02)00011-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cloned T9 glioma cells (T9-C2) expressing the membrane form of macrophage colony stimulating factor (mM-CSF) inoculated subcutaneously into rats do not grow and glioma-specific immunity is stimulated. Immunotherapy experiments showed that intracranial T9 tumors present for one to four days could be successfully eradicated by peripheral vaccination with T9-C2 cells. CD4+ and CD8+ T splenocytes from immunized rats, when restimulated in vitro with T9 cells, produced interleukin-2 and -4. Protective immunity against intracranial T9 gliomas could only be adoptively transferred into naive rats by the CD4+ splenocytes obtained from T9-C2 immunized rats. Rats immunized by the T9-C2 tumor cells also resisted two different syngeneic gliomas (RT2 and F98) but allowed a syngeneic NUTU-19 ovarian cancer to grow. Such cross-protective immunity against unrelated gliomas suggests that mM-CSF transfected tumor cells have immunotherapeutic potential for use as an allogeneic tumor vaccine.
Collapse
Affiliation(s)
- Ramon Sanchez
- Diagnostic and Molecular Health Care Group, Box 113 Veterans Affairs Medical Center, 5901 E. 7th Street, Long Beach, CA 90822, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Anderson RC, Elder JB, Brown MD, Mandigo CE, Parsa AT, Kim PD, Senatus P, Anderson DE, Bruce JN. Changes in the immunologic phenotype of human malignant glioma cells after passaging in vitro. Clin Immunol 2002; 102:84-95. [PMID: 11781071 DOI: 10.1006/clim.2001.5152] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although immunotherapeutic strategies against glioblastomas have been promising both in vitro and in animal models, similar successes have not been realized in human clinical trials. One reason may be that immunotherapeutic strategies are based on prior studies that primarily have used human glioblastoma cell lines passaged in vitro, which may not accurately reflect the in vivo properties of glioblastoma cells. In this report, we used flow cytometry to quantify the expression of immunological cell surface molecules on human glioblastomas directly ex vivo (prior to any in vitro culturing) and after varying passages in vitro. Furthermore, we used ELISA to quantitate cytokine secretion after various passages in vitro. We demonstrate that in vitro culturing of established cell lines led to increases in the cell surface expression of MHC class I and ICAM-1 and secretion of IL-6 and TGF-beta(2). Furthermore, there were significant changes in the expression of MHC class I, MHC class II, B7-2, ICAM-1, and FasL when comparing ex vivo tumor cells to those after a single passage in vitro. After passaging once in vitro, there were also significant changes in the secretion of TGF-beta(2) and IL-10. This report indicates that in vitro culturing leads to significant changes in both cell surface molecules and secreted cytokines, which are known to affect the ability of immune cells to initiate an anti-tumor immune response. These changes in the immunological phenotype of glioblastomas after in vitro culturing may in part explain the limited success of immunotherapeutic strategies against glioblastomas in human clinical trials.
Collapse
Affiliation(s)
- Richard C Anderson
- Department of Neurological Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Soos JM, Krieger JI, Stüve O, King CL, Patarroyo JC, Aldape K, Wosik K, Slavin AJ, Nelson PA, Antel JP, Zamvil SS. Malignant glioma cells use MHC class II transactivator (CIITA) promoters III and IV to direct IFN-gamma-inducible CIITA expression and can function as nonprofessional antigen presenting cells in endocytic processing and CD4(+) T-cell activation. Glia 2001; 36:391-405. [PMID: 11746775 DOI: 10.1002/glia.1125] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Malignant gliomas (MGs), lethal human central nervous system (CNS) neoplasms, contain tumor infiltrating lymphocytes (TIL). Although MHC class II molecules are frequently detected on MG cells, suggesting that they may be capable of antigen (Ag) presentation to CD4(+) T cells, deficiencies in CD4(+) T-cell activation are associated with these nonimmunogenic tumors. We evaluated regulation of the MHC class II transactivator (CIITA), the key intermediate that controls class II expression, in MG cells and tested whether MG cells could process native Ag. After interferon-gamma (IFN-gamma) stimulation, MG cells upregulated CIITA and class II molecules. IFN-gamma-inducible CIITA expression in MG cells, as well as primary human astrocytes, was directed by two CIITA promoters, pIV, the promoter for IFN-gamma-inducible CIITA expression in nonprofessional antigen-presenting cells (APC), and pIII, the promoter that directs constitutive CIITA expression in B cells. Both pIII and pIV directed CIITA transcription in vivo in MGs and ex vivo in IFN-gamma-activated primary MG cultures. We also demonstrate for the first time that MG cells can process native Ag for presentation to CD4(+) MHC class II-restricted Th1 cells, indicating that MG cells can serve as nonprofessional APC. CIITA may be a key target to modulate MHC class II expression, which could augment immunogenicity, Ag presentation, and CD4(+) T-cell activation in MG therapy.
Collapse
MESH Headings
- Adult
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Astrocytes/cytology
- Astrocytes/immunology
- Astrocytes/metabolism
- Autoantigens/immunology
- Autoantigens/pharmacology
- Base Sequence/genetics
- Brain Neoplasms/immunology
- Brain Neoplasms/metabolism
- Brain Neoplasms/physiopathology
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Exons/genetics
- Exons/immunology
- Female
- Gene Expression Regulation, Neoplastic/genetics
- Gene Expression Regulation, Neoplastic/immunology
- Glioma/immunology
- Glioma/metabolism
- Glioma/physiopathology
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Immunohistochemistry
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Male
- Middle Aged
- Molecular Sequence Data
- Myelin Basic Protein/immunology
- Myelin Basic Protein/pharmacology
- Nuclear Proteins
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/immunology
- RNA, Messenger/immunology
- RNA, Messenger/metabolism
- Trans-Activators/genetics
- Trans-Activators/immunology
- Trans-Activators/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J M Soos
- Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Basse PH, Whiteside TL, Chambers W, Herberman RB. Therapeutic activity of NK cells against tumors. Int Rev Immunol 2001; 20:439-501. [PMID: 11878512 DOI: 10.3109/08830180109054416] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
While it is generally accepted that natural killer (NK) cells, by killing tumor cells in the circulation, represent a first line of defense against metastases, their therapeutic activity against established tumors has been limited. In this review, we describe studies to improve the therapeutic effectiveness of activated NK cells in both animal models and clinical trials to better understand the biological problems that limit their effectiveness.
Collapse
|
36
|
Schneider T, Gerhards R, Kirches E, Firsching R. Preliminary results of active specific immunization with modified tumor cell vaccine in glioblastoma multiforme. J Neurooncol 2001; 53:39-46. [PMID: 11678429 DOI: 10.1023/a:1011856406683] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECT Treatment for glioblastoma multiforme has failed to show any progress for decades. While specific immunization with tumor cells modified with Newcastle-Disease-Virus (NDV) has been reported successful in some extracerebral tumors, its effect on glioblastoma is unknown. We report on 11 patients, in whom this approach was analyzed. METHODS A vaccine was produced from autologous tumor cell cultures of 11 patients with glioblastoma. After completed surgery and radiotherapy an intracutaneous vaccination was performed 4 times with a 2 week interval and finally after 3 months. Local reactions, general side effects and survival were monitored closely. RESULTS The local reaction of the skin after injection of vaccine increased from 1.67 to 4.05 cm2 in 8 weeks. The skin reaction after parallel injection of inactivated, untreated tumor cells increased from 0.11 to 1.09 cm2. The median survival was 46 weeks (mean 60 weeks). No side effects were noted. CONCLUSION Active specific immunization with NDV-modified glioblastoma cells produced a noticeable peripheral immune response. In this preliminary series survival of patients was not significantly longer after active specific immunization than after combined treatment of surgery, radiotherapy and chemotherapy. As there were no side effects, however, active specific immunization may be considered an alternative in the management of glioblastoma.
Collapse
Affiliation(s)
- T Schneider
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Germany
| | | | | | | |
Collapse
|
37
|
Sloan AE, Dansey R, Zamorano L, Barger G, Hamm C, Diaz F, Baynes R, Wood G. Adoptive immunotherapy in patients with recurrent malignant glioma: preliminary results of using autologous whole-tumor vaccine plus granulocyte-macrophage colony–stimulating factor and adoptive transfer of anti-CD3–activated lymphocytes. Neurosurg Focus 2000; 9:e9. [PMID: 16817692 DOI: 10.3171/foc.2000.9.6.10] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
This trial was designed to determine the ability of autologous whole–tumor cell vaccines to induce cell-mediated immune responses in patients with recurrent malignant glioma, as well as to determine whether combining such vaccination with adoptive transfer of in vitro activated T lymphocytes prolongs patient survival.
Methods
Nineteen patients with recurrent malignant glioma, in whom previous external beam radiotherapy and at least one course of chemotherapy had failed were vaccinated twice with irradiated autologous whole tumor cells by using granulocyte-marcrophage colony–stimulating factor as an adjuvant. Patients then underwent leukapheresis followed by adoptive transfer of peripheral blood lymphocytes activated in vitro with anti-CD3 and interleukin-2. In vivo immune response, radiological response, clinical outcome, and survival were monitored.
Seventeen patients developed a delayed-type hypersensitivity (DTH) response to vaccination that appeared to be directed against the autologous tumor. In eight patients there was radiological evidence of a response and in five there was evidence of clinical improvement. Median survival was 12 months (range 6–28 months), and both the presence of a DTH response and the radiological response correlated with survival (p < 0.02 and p < 0.04, respectively).
Conclusions
These preliminary results suggest that autologous whole–tumor cell vaccines induce a cell-mediated immune response, which appears to be tumor specific in most patients. Furthermore, vaccination combined with adoptive immunotherapy with in vitro activated cells may induce a radiologically demonstrated tumor response and improved survival despite a condition of advanced disease and immunosuppression resulting from previous treatment or tumor burden. Further studies of immunotherapy are warranted.
Collapse
Affiliation(s)
- A E Sloan
- Department of Neurosurgery, Karmanos Cancer Institute, Detroit, Michigan, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Pollack IF, Okada H, Chambers WH. Exploitation of immune mechanisms in the treatment of central nervous system cancer. Semin Pediatr Neurol 2000; 7:131-43. [PMID: 10914414 DOI: 10.1053/pb.2000.6691] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Malignant gliomas are among the most common intrinsic brain tumors of both children and adults, and, because of unique aspects of their biology and anatomic site, they are the most refractory to conventional therapeutic strategies involving surgery, radiotherapy, or chemotherapy. Given the failure of standard therapies to improve the outlook of affected patients, significant attention has been focused on development of alternative treatments, particularly immunotherapy. Attempts have been made to treat gliomas using a variety of immunologically based strategies, including passive immunization, adoptive cellular immunotherapy, local and systemic delivery of biological response modifiers, and vaccination with tumor cells. Although preclinical modeling of these therapies provided an impetus for translation of their results into clinical protocols, these therapies have failed to yield consistently promising results in initial trials. However, significant insights into the immunobiology of the central nervous system (CNS) and gliomas have been gained from these studies, and have established that a number of immunobiological features of the brain and of gliomas themselves may be critical determinants in regulating efficacious treatment of these tumors. These include the following: (1) the presence of a blood-brain barrier that, although partially disrupted by the tumor, functions to exclude elements of the immune system from the tumor or brain parenchyma; (2) a lack of organized secondary lymphatic tissues supporting efficient immune responses locally in the CNS; (3) low levels of expression of major histocompatibility complex proteins in the CNS; (4) an apparent paucity of the most efficient antigen-presenting cells; and (5) glioma-derived immunosuppressive factors, such as transforming growth factor-beta, that interfere with the induction of local as well as systemic immune responses to the tumor. Recognition of these factors, and an appreciation of the underlying need for and validity of developing immunologically based therapies for gliomas, supports continued development of novel immunotherapeutic approaches, particularly those attempting to enhance the immunogenicity of glioma cells. This review addresses the current state of knowledge regarding the immunobiology of gliomas, recent developments in immunotherapy of gliomas, and promising future directions for development and implementation of cellular immunotherapy of gliomas.
Collapse
Affiliation(s)
- I F Pollack
- Department of Neurosurgery, University of Pittsburgh Cancer Institute Brain Tumor Center, Children's Hospital of Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
39
|
Quattrocchi KB, Miller CH, Cush S, Bernard SA, Dull ST, Smith M, Gudeman S, Varia MA. Pilot study of local autologous tumor infiltrating lymphocytes for the treatment of recurrent malignant gliomas. J Neurooncol 2000; 45:141-57. [PMID: 10778730 DOI: 10.1023/a:1006293606710] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A prospective pilot study was performed in order to assess the safety of treating recurrent malignant gliomas (MGs) with locally infused autologous tumor infiltrating lymphocytes (TILs) and recombinant interleukin-2 (rIL-2). Six patients were entered between June 27, 1994 and June 2, 1995 and followed until July 1, 1998. At surgery an Ommaya reservoir was placed for later infusion of TILs and rIL-2. Following surgery, autologous TILs were expanded in vitro in the presence of rIL-2 and infused on treatment days 1 and 14, with concurrent rIL-2 infusions performed three times each week for one month. Following completion of immunotherapy all patients were offered chemotherapy. Phenotypic analysis demonstrated TILs to be T-lymphocytes (87-99% CD3+). Of these, 4 of 6 cases (67%) phenotyped as cytotoxic/suppressor T-lymphocytes (CD8+) and 2 of 6 cases (33%) phenotyped as helper/inducer T-lymphocytes (CD4+). TILs demonstrated limited selective cytotoxicity, with dose dependent cytotoxicity against autologous tumor, allogenic tumor and long term MG cell lines. There were no significant (Grade 3 or 4) complications. One patient developed transient low grade fevers, and 2 developed asymptomatic hydrocephalus. All patients developed transient and asymptomatic cerebral swelling, noted on the immediate post-treatment imaging studies. At three and six month follow-up, 3 patients responded with partial response, 2 demonstrated stable disease and 1 patient progressed. At long term follow-up, 1 patient had a complete response (45 month follow-up), 2 had a partial response (48 and 47 month follow-up) and 3 patients expired as a result of progressive disease (at 12, 12 and 18 months following immunotherapy). A relationship between subsequent chemotherapy or extent of resection to outcome was not apparent but could not be excluded. This pilot study demonstrated that locally infused autologous TILs and rIL-2 could be delivered without serious toxicity. Further studies are indicated to determine the safety and long term efficacy of TIL immunotherapy.
Collapse
Affiliation(s)
- K B Quattrocchi
- Division of Neurosurgery, University of North Carolina, Chapel Hill, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Abstract
OBJECTIVE Despite advances in conventional therapy, the prognosis for most glioma patients remains dismal. This has prompted an intensive search for effective treatment alternatives. Immunotherapy, one such alternative, has long been recognized as a potentially potent cancer treatment but has been limited by an inadequate understanding of the immune system. Now, increased insight into immunology is suggesting more rational approaches to immunotherapy. In this article, we explore key aspects of modern immunology and discuss their implications for glioma therapy. METHODS A thorough literature review of glioma immunology and immunotherapy was undertaken to inquire into the basic immunology, central nervous system immunology, glioma immunobiology, standard glioma immunotherapy, and recent immunotherapeutic advances in glioma treatment. RESULTS Although gliomas express tumor-associated antigens and appear potentially sensitive to immune responses, many factors work together to inhibit antiglioma immunity. Not surprisingly, most clinical attempts at glioma immunotherapy have met with little success to date. However, novel immunostimulatory strategies, such as immunogene therapy, directed cytokine delivery, and dendritic cell manipulation, have recently yielded dramatic preclinical results in glioma models. This suggests that glioma-derived immunosuppression can be overcome. CONCLUSION Modern molecular biology and immunology techniques have yielded a wealth of new data about glioma immunobiology. Armed with this information, many investigators have proposed novel means to stimulate antiglioma immune responses. Although definitive clinical results remain to be seen, the current renaissance in glioma immunology and immunotherapy shows great promise for the future.
Collapse
Affiliation(s)
- I F Parney
- Department of Surgery, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
42
|
Hegardt P, Widegren B, Sjögren HO. Nitric-oxide-dependent systemic immunosuppression in animals with progressively growing malignant gliomas. Cell Immunol 2000; 200:116-27. [PMID: 10753503 DOI: 10.1006/cimm.2000.1625] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of nitric oxide (NO) and adherent spleen cells in systemic immunosuppression developing in animals carrying malignant glioma isografts was analyzed. Rats harboring a subcutaneous glioma isograft for 3 weeks were immunized with glioma cells genetically engineered to express IFN-gamma. One week later spleen cells were tested for immune responsiveness in vitro. A decreased cytotoxic activity of NK-cells and T-cells compared to tumor-free animals immunized in parallel was shown. Spleen cell proliferative responses to tumor cells, SEA, and anti-CD3 were all significantly suppressed, as was the production of IFN-gamma and IL-10. Plastic adherent spleen cells from tumor-bearing rats suppressed the SEA-induced proliferative response and the production of IFN-gamma and IL-10 by nonadherent spleen cells from tumor-free rats. A major part of this suppression appears to be dependent on the production of NO because suppression was efficiently counteracted in vitro by the NO-synthase inhibitor N-nitro-l-arginine methyl ester. Moreover, a significantly increased level of nitrite in culture supernatants correlated with the observed suppression. We conclude that the systemic immunosuppression associated with growing gliomas is in part mediated by mechanisms dependent on NO overproduction in adherent spleen cells.
Collapse
Affiliation(s)
- P Hegardt
- Department of Cell and Molecular Biology, Section for Tumor Immunology, Immunology, Sölvegatan 19, Lund, S-223 62, Sweden
| | | | | |
Collapse
|
43
|
Sablotzki A, Ebel H, Mühling J, Dehne MG, Nopens H, Giesselmann H, Hempelmann G. Dysregulation of immune response following neurosurgical operations. Acta Anaesthesiol Scand 2000; 44:82-7. [PMID: 10669277 DOI: 10.1034/j.1399-6576.2000.440115.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Postoperative infections are common and potentially fatal complications in neurosurgical intensive care medicine. An impairment of immune function has been described after central nervous system surgery and in patients harboring malignant brain tumors. The aim of our study was to investigate whether differences in cell-mediated immunity can be found in patients undergoing craniotomy for surgery of glioblastoma or clipping of an intracerebral aneurysm. METHODS In order to determine the influence of the underlying disease on the immune system, we measured changes in cytokine concentrations (IL-6, IL-10, TGF-beta1) and lymphocyte-subsets (CD3+, CD3+HLA-DR+, CD4+, CD8+, CD19+, and CD16+56+) in 8 patients with glioblastoma and in 8 patients with an intracerebral aneurysm before, during and after the neurosurgical procedure. RESULTS In the comparison of glioblastoma and aneurysm patients, we could show that IL-6 plasma levels were pre- and intraoperatively higher in the aneurysm-group (P<0.05), and the plasma concentrations of IL-10 and TGF-beta were significantly elevated in the glioma-group. The lymphocyte-subsets showed a significantly lower percentage of NK-cells and activated T-cells in the glioma-group. CONCLUSION Our results document a significant dysregulation of immune response in glioma patients. This may be induced by elevated plasma concentrations of immunoinhibiting cytokines IL-10 and transforming growth factor-beta 1.
Collapse
Affiliation(s)
- A Sablotzki
- Department of Anaesthesiology and Intensive Care Medicine, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
de Tribolet N, Campiche R. Neurosurgery at the University of Lausanne. Neurosurgery 1999; 45:901-6. [PMID: 10515487 DOI: 10.1097/00006123-199910000-00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The University of Lausanne was founded in 1537. The faculty of medicine was created in 1890, and the service of surgery was directed by César Roux. Roux, a well-known surgeon, was visited by Harvey Cushing during 1900-1901. In the early 1930s, Jean Rossier from Lausanne trained with Cushing, but Rossier passed away in 1942. Eric Zander created the division of neurosurgery in 1959; it became an independent service in 1967. Nicolas de Tribolet served as chairman from 1984 until 1994, when he was asked to take charge of the merger of the university services of Geneva and Lausanne. In October 1997, Jean-Guy Villemure joined him in the newly merged department, becoming chairman in Lausanne, while de Tribolet is chairman in Geneva and head of the department comprising both services.
Collapse
Affiliation(s)
- N de Tribolet
- Department de Neurochirurgie, Hôpital Cantonal Universitaire, Geneva, Switzerland
| | | |
Collapse
|
45
|
Abstract
Since the declaration of the war on cancer in 1971, our ability to effectively treat cancer has been less successful than anticipated. Surgery and radiation therapy remain our most effective treatment modalities, with chemotherapy proving beneficial in only a limited number of tumor types. The reality of this poor response to conventional therapy has prompted a search for other potentially beneficial therapies. The idea of using the immune system to eradicate tumor is not new. Over 100 years ago, William Coley (in 1893) first reported on the ability to induce tumor regressions by nonspecific activation of the immune system in response to bacterial toxins. Despite this early beginning, efforts to reliably manipulate the immune system to promote tumor regression has been universally disappointing. With recent advances in our understanding of the immune system, and the identification and availability of numerous growth promoting and growth-suppression cytokines, the concept of immunotherapy being a useful therapeutic intervention for the treatment of cancer is becoming a reality. Immunology in general, and tumor immunology specifically, are fields foreign to the practicing tumor surgeon. As progress in these fields are made, it will become important for the surgical oncologist to have a better understanding of tumor immunology as it relates to therapy. This paper reviews our current understanding of the immune system as it relates to cancer immunotherapy (using primary intracranial glioma as the tumor model), and then relates this knowledge to recent work in the development of tumor-specific vaccines.
Collapse
Affiliation(s)
- K O Lillehei
- Department of Surgery, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | |
Collapse
|
46
|
Perrin G, Schnuriger V, Quiquerez AL, Saas P, Pannetier C, de Tribolet N, Tiercy JM, Aubry JP, Dietrich PY, Walker PR. Astrocytoma infiltrating lymphocytes include major T cell clonal expansions confined to the CD8 subset. Int Immunol 1999; 11:1337-50. [PMID: 10421791 DOI: 10.1093/intimm/11.8.1337] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Anaplastic astrocytoma and glioblastoma are frequent and malignant brain tumors that are infiltrated by T lymphocytes. Whether these cells result from non-specific inflammation following blood-brain barrier disruption or an antigen-driven specific immune response is unknown. In this study, an in-depth characterization of TCR diversity in tumor and blood RNA biopsies was performed in a series of 16 patients with malignant astrocytoma. Whilst there was no obvious restriction of the AV and BV gene segment usage, complementarity-determining region 3 size analysis and sequencing of amplified TCR transcripts revealed multiple T cell oligoclonal expansions in all astrocytomas analyzed. Unique T cell clones were present in different adjacent areas of a given tumor, but never detected in the blood. Quantification of the number of TCR clonal transcripts per microg of tumor RNA indicated that certain T cell clonal expansions may represent at least 300 cells/10(6) tumor cells. Furthermore, we demonstrated that the in vivo expanded clones were almost exclusively confined to the CD8(+) subset. Overall, these data suggest that spontaneous antigen-driven immune responses may be elicited against human astrocytoma despite the immunosuppressive microenvironment generated by the brain and the tumor itself. However, the ultimate failure of the immune system to control tumor growth could be the consequence of a deficient CD4 T(h) component of the response. This observation could have important consequences for the development of immunotherapies for astrocytoma patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Amino Acid Sequence
- Astrocytoma/immunology
- Brain Neoplasms/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Clone Cells
- Complementarity Determining Regions
- Female
- Genes, T-Cell Receptor
- Humans
- Immunoglobulin Variable Region/genetics
- Lymphocytes, Tumor-Infiltrating/cytology
- Lymphocytes, Tumor-Infiltrating/immunology
- Male
- Middle Aged
- Molecular Sequence Data
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- G Perrin
- Laboratory of Tumor Immunology, Division of Oncology, University Hospital, 1211 Geneva 14, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
BACKGROUND Bacille Calmette-Guérin (BCG), an attenuated strain of tuberculous bacillus, is the source of vaccines providing unclear and variable protection against tuberculosis (TB) and cancer. Thermostable macromolecular antigens (TMAs) are major mycobacterial complexes immunodominant in disease. A60 (TMA complex of BCG) protects mice against TB development, via T lymphocyte (TL)-mediated macrophage (Mphi) activation, halting intracellular mycobacterial replication. In most A60-primed mice, cytolytic TLs and Mphi infiltrate cancer tissue, resulting in 80-100% rejection. Adoptive TL transfer is indispensable for Mphi-dependent tumour cell inactivation via oxygen and nitrogen radicals. Neoplasm development induces immune anergy with depletion ofA60-specific TL and activated Mphi. A60 protects mice against TB and cancer by inducing the synthesis of three lymphokines: interleukin 2 (IL-2), interferon gamma (IFN-gamma) and tumour necrosis factor alpha (TNF-alpha). Tumour cells prevent A60-dependent synthesis of these lymphokines in vivo and in vitro. CONCLUSION These data provide some clues to immune surveillance and tumour escape mechanisms, as well as to the antituberculous and antineoplastic BCG action.
Collapse
Affiliation(s)
- C Cocito
- Microbiology and Molecular Genetics Laboratory, University of Louvain Medical School, Brussels, Belgium
| | | |
Collapse
|
48
|
Abstract
The study of cellular adhesion molecules offers crucial understanding of cellular interactions. Their name implies an underestimation of their function, as intercellular glue. In fact, they play vital roles in tissue development and intra- and intercellular signaling. In neurology, cellular adhesion molecules are already providing welcome new insight into neurodevelopmental anomalies, autoimmune demyelination, and invasive tumours. Cellular adhesion molecule manipulation has led to several therapeutic options which are the subject of ongoing clinical investigation.
Collapse
Affiliation(s)
- P J Marchetti
- Division of Neurology, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | | |
Collapse
|
49
|
Maes H, Cocito C. In vitro analysis of cancer prevention by a mycobacterial antigen complex and of cancer-promoted inhibition of immune reactions. J Cancer Res Clin Oncol 1996; 122:727-34. [PMID: 8954170 DOI: 10.1007/bf01209120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The antigen complex A60 of Mycobacterium bovis bacillus Calmette-Guérin protected mice against experimental tuberculous infection, and prevented cancer development after challenge with EMT 6 cells. Although humoral and cellular immune reactions elicited by A60 in vivo remained unaffected in cases of tumor rejection, they were suppressed in the case of neoplastic growth. In the present work, these in vivo observations were analyzed by in vitro techniques. Activated macrophages played a major role, and cytolytic T lymphocytes a minor role, in A60-promoted cancer cell cytolysis leading to tumor rejection. In vitro, EMT 6 cells weakly inhibited the proliferation of A60-specific B lymphocytes and strongly inhibited the functions of activated macrophages. However, the collapse of both humoral and cellular immune reactions during the course of cancer development was also accompanied by an inhibitory action of EMT 6 cells on the multiplication and functions of A60-specific T lymphocytes. Tumor-dependent repression of macrophage activation was therefore due to both a direct action of tumor cells on macrophages and an indirect one via inhibition of macrophage-activating T cell functions. On the other hand, tumor-induced collapse of the anti-A60 Ig synthesis was mainly due to inhibition of B-cell-activating T cells, with a weaker direct effect of tumor cells on B lymphocytes. Consequently, A60 and tumor cells exert opposite effects on the immune system at several levels.
Collapse
Affiliation(s)
- H Maes
- Microbiology and Molecular Genetics Section, University of Louvain, GEMO-UCL 5225, Brussels, Belgium
| | | |
Collapse
|
50
|
Giometto B, Bozza F, Faresin F, Alessio L, Mingrino S, Tavolato B. Immune infiltrates and cytokines in gliomas. Acta Neurochir (Wien) 1996; 138:50-6. [PMID: 8686525 DOI: 10.1007/bf01411724] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Frozen sections of 21 gliomas were analysed to characterize inflammatory infiltrating cells, HLA-DR antigen expression and cytokine secretion. Mononuclear cells infiltrating the tumours were mostly macrophages, which were detected in 100% of cases, and expressed HLA-DR antigens. Lymphocytes were less frequently seen and expressed the CD8 phenotype. Interleukin-1 beta (IL-1 beta) and Interleukin-6 (IL-6), two cytokines mainly produced by activated cells of the macrophage lineage, were demonstrated especially in neoplastic astrocytes. IL-1 beta immunoreactivity was detected in all tumours, and was prevalent in more anaplastic gliomas; IL-6 was found in anaplastic gliomas and in glioblastomas. IL-1 receptors were expressed by both infiltrating macrophages and neoplastic astrocytes in the gliomas analysed. These findings suggest that cytokine production in gliomas seems not related to immune reactions against the tumour and their synthesis by anaplastic astrocytes could follow an unregulated activation of many metabolic processes after neoplastic transformation.
Collapse
Affiliation(s)
- B Giometto
- Institute of Neurology (Second Clinic), University of Padua, School of Medicine, Italy
| | | | | | | | | | | |
Collapse
|