51
|
Gardeck AM, Sheehan J, Low WC. Immune and viral therapies for malignant primary brain tumors. Expert Opin Biol Ther 2017; 17:457-474. [DOI: 10.1080/14712598.2017.1296132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andrew M. Gardeck
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Sheehan
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
52
|
Abstract
Glioblastoma Multiforme (GBM) is the most common malignant primary brain neoplasm having a mean survival time of <24 months. This figure remains constant, despite significant progress in medical research and treatment. The lack of an efficient anti-tumor immune response and the micro-invasive nature of the glioma malignant cells have been explained by a multitude of immune-suppressive mechanisms, proven in different models. These immune-resistant capabilities of the tumor result in a complex interplay this tumor shares with the immune system. We present a short review on the immunology of GBM, discussing the different unique pathological and molecular features of GBM, current treatment modalities, the principles of cancer immunotherapy and the link between GBM and melanoma. Current knowledge on immunological features of GBM, as well as immunotherapy past and current clinical trials, is discussed in an attempt to broadly present the complex and formidable challenges posed by GBM.
Collapse
|
53
|
Choy W, Lagman C, Lee SJ, Bui TT, Safaee M, Yang I. Impact of Human Immunodeficiency Virus in the Pathogenesis and Outcome of Patients with Glioblastoma Multiforme. Brain Tumor Res Treat 2016; 4:77-86. [PMID: 27867916 PMCID: PMC5114196 DOI: 10.14791/btrt.2016.4.2.77] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/10/2016] [Accepted: 07/18/2016] [Indexed: 12/25/2022] Open
Abstract
Background Improvement in antiviral therapies have been accompanied by an increased frequency of non-Acquired Immune Deficiency Syndrome (AIDS) defining malignancies, such as glioblastoma multiforme. Here, we investigated all reported cases of human immunodeficiency virus (HIV)-positive patients with glioblastoma and evaluated their clinical outcomes. A comprehensive review of the molecular pathogenetic mechanisms underlying glioblastoma development in the setting of HIV/AIDS is provided. Methods We performed a PubMed search using keywords “HIV glioma” AND “glioblastoma,” and “AIDS glioma” AND “glioblastoma.” Case reports and series describing HIV-positive patients with glioblastoma (histologically-proven World Health Organization grade IV astrocytoma) and reporting on HAART treatment status, clinical follow-up, and overall survival (OS), were included for the purposes of quantitative synthesis. Patients without clinical follow-up data or OS were excluded. Remaining articles were assessed for data extraction eligibility. Results A total of 17 patients met our inclusion criteria. Of these patients, 14 (82.4%) were male and 3 (17.6%) were female, with a mean age of 39.5±9.2 years (range 19–60 years). Average CD4 count at diagnosis of glioblastoma was 358.9±193.4 cells/mm3. Tumor progression rather than AIDS-associated complications dictated patient survival. There was a trend towards increased median survival with HAART treatment (12.0 vs 7.5 months, p=0.10) Conclusion Our data suggests that HAART is associated with improved survival in patients with HIV-associated glioblastoma, although the precise mechanisms underlying this improvement remain unclear.
Collapse
Affiliation(s)
- Winward Choy
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Carlito Lagman
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Seung J Lee
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Timothy T Bui
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Safaee
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Isaac Yang
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA.; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
54
|
Chimeric antigen receptors for treatment of glioblastoma: a practical review of challenges and ways to overcome them. Cancer Gene Ther 2016; 24:121-129. [PMID: 27767090 DOI: 10.1038/cgt.2016.46] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is by far the most common and the most aggressive of all the primary brain malignancies. No curative therapy exists, and median life expectancy hovers at around 1 year after diagnosis, with a minute fraction surviving beyond 5 years. The difficulty in treating GBM lies in the cancer's protected niche within the blood-brain barrier and the heterogeneity of the cancer cells, which possess varying degrees of susceptibility to various common modalities of treatment. Over time, it is the tumor heterogeneity of GBM and the ability of the cancer stem cells to evolve in response treatment that renders the cancer refractory to conventional treatment. Therefore, research has increasingly focused on treatment that incorporates knowledge of GBM molecular biology to therapeutic strategies. One type of therapy that shows great promise is the area of T-cell immunotherapy to target GBM-specific tumor antigens. One attractive strategy is to use T cells that have undergone genetic modification to express a chimeric antigen receptor capable of interacting with tumor antigens. In this article, we will review chimeric antigen receptor T-cell therapy, their advantages, drawbacks, challenges facing their use and how those challenges may be overcome.
Collapse
|
55
|
Kamran N, Calinescu A, Candolfi M, Chandran M, Mineharu Y, Asad AS, Koschmann C, Nunez FJ, Lowenstein PR, Castro MG. Recent advances and future of immunotherapy for glioblastoma. Expert Opin Biol Ther 2016; 16:1245-64. [PMID: 27411023 PMCID: PMC5014608 DOI: 10.1080/14712598.2016.1212012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/08/2016] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Outcome for glioma (GBM) remains dismal despite advances in therapeutic interventions including chemotherapy, radiotherapy and surgical resection. The overall survival benefit observed with immunotherapies in cancers such as melanoma and prostate cancer has fuelled research into evaluating immunotherapies for GBM. AREAS COVERED Preclinical studies have brought a wealth of information for improving the prognosis of GBM and multiple clinical studies are evaluating a wide array of immunotherapies for GBM patients. This review highlights advances in the development of immunotherapeutic approaches. We discuss the strategies and outcomes of active and passive immunotherapies for GBM including vaccination strategies, gene therapy, check point blockade and adoptive T cell therapies. We also focus on immunoediting and tumor neoantigens that can impact the efficacy of immunotherapies. EXPERT OPINION Encouraging results have been observed with immunotherapeutic strategies; some clinical trials are reaching phase III. Significant progress has been made in unraveling the molecular and genetic heterogeneity of GBM and its implications to disease prognosis. There is now consensus related to the critical need to incorporate tumor heterogeneity into the design of therapeutic approaches. Recent data also indicates that an efficacious treatment strategy will need to be combinatorial and personalized to the tumor genetic signature.
Collapse
Affiliation(s)
- Neha Kamran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Alexandra Calinescu
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Marianela Candolfi
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Mayuri Chandran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Yohei Mineharu
- d Department of Neurosurgery , Kyoto University Graduate School of Medicine , Kyoto , Japan
| | - Antonela S Asad
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Carl Koschmann
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Felipe J Nunez
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Pedro R Lowenstein
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Maria G Castro
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| |
Collapse
|
56
|
Yaghi L, Poras I, Simoes RT, Donadi EA, Tost J, Daunay A, de Almeida BS, Carosella ED, Moreau P. Hypoxia inducible factor-1 mediates the expression of the immune checkpoint HLA-G in glioma cells through hypoxia response element located in exon 2. Oncotarget 2016; 7:63690-63707. [PMID: 27577073 PMCID: PMC5325396 DOI: 10.18632/oncotarget.11628] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/20/2016] [Indexed: 12/31/2022] Open
Abstract
HLA-G is an immune checkpoint molecule with specific relevance in cancer immunotherapy. It was first identified in cytotrophoblasts, protecting the fetus from maternal rejection. HLA-G tissue expression is very restricted but induced in numerous malignant tumors such as glioblastoma, contributing to their immune escape. Hypoxia occurs during placenta and tumor development and was shown to activate HLA-G. We aimed to elucidate the mechanisms of HLA-G activation under conditions combining hypoxia-mimicking treatment and 5-aza-2'deoxycytidine, a DNA demethylating agent used in anti-cancer therapy which also induces HLA-G. Both treatments enhanced the amount of HLA-G mRNA and protein in HLA-G negative U251MG glioma cells. Electrophoretic Mobility Shift Assays and luciferase reporter gene assays revealed that HLA-G upregulation depends on Hypoxia Inducible Factor-1 (HIF-1) and a hypoxia responsive element (HRE) located in exon 2. A polymorphic HRE at -966 bp in the 5'UT region may modulate the magnitude of the response mediated by the exon 2 HRE. We suggest that therapeutic strategies should take into account that HLA-G expression in response to hypoxic tumor environment is dependent on HLA-G gene polymorphism and DNA methylation state at the HLA-G locus.
Collapse
Affiliation(s)
- Layale Yaghi
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR E5, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
- Lebanese University, School of Medicine, Hadath, Lebanon
| | - Isabelle Poras
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR E5, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Renata T. Simoes
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR E5, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
- Instituto de Ensino e Pesquisa da Santa Casa de Belo Horizonte, IEP/SCBH, Belo Horizonte, Minas Gerais, Brasil
| | - Eduardo A. Donadi
- Divisão de Imunologia Clínica, Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Jörg Tost
- Centre d'Etude du Polymorphisme Humain, Fondation Jean-Dausset, Laboratory for Functional Genomics, Paris, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Genotypage, Laboratory for Epigenetics and Environment, Evry, France
| | - Antoine Daunay
- Centre d'Etude du Polymorphisme Humain, Fondation Jean-Dausset, Laboratory for Functional Genomics, Paris, France
| | - Bibiana Sgorla de Almeida
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR E5, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
- Divisão de Imunologia Clínica, Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Edgardo D. Carosella
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR E5, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Philippe Moreau
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- Université Paris-Diderot, Sorbonne Paris-Cité, UMR E5, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
57
|
Zhang L, Ren J, Zhang H, Cheng G, Xu Y, Yang S, Dong C, Fang D, Zhang J, Yang A. HER2-targeted recombinant protein immuno-caspase-6 effectively induces apoptosis in HER2-overexpressing GBM cells in vitro and in vivo. Oncol Rep 2016; 36:2689-2696. [DOI: 10.3892/or.2016.5088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 07/18/2016] [Indexed: 11/05/2022] Open
|
58
|
Hegde M, Mukherjee M, Grada Z, Pignata A, Landi D, Navai SA, Wakefield A, Fousek K, Bielamowicz K, Chow KK, Brawley VS, Byrd TT, Krebs S, Gottschalk S, Wels WS, Baker ML, Dotti G, Mamonkin M, Brenner MK, Orange JS, Ahmed N. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest 2016; 126:3036-52. [PMID: 27427982 PMCID: PMC4966331 DOI: 10.1172/jci83416] [Citation(s) in RCA: 498] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 05/13/2016] [Indexed: 12/24/2022] Open
Abstract
In preclinical models of glioblastoma, antigen escape variants can lead to tumor recurrence after treatment with CAR T cells that are redirected to single tumor antigens. Given the heterogeneous expression of antigens on glioblastomas, we hypothesized that a bispecific CAR molecule would mitigate antigen escape and improve the antitumor activity of T cells. Here, we created a CAR that joins a HER2-binding scFv and an IL13Rα2-binding IL-13 mutein to make a tandem CAR exodomain (TanCAR) and a CD28.ζ endodomain. We determined that patient TanCAR T cells showed distinct binding to HER2 or IL13Rα2 and had the capability to lyse autologous glioblastoma. TanCAR T cells exhibited activation dynamics that were comparable to those of single CAR T cells upon encounter of HER2 or IL13Rα2. We observed that TanCARs engaged HER2 and IL13Rα2 simultaneously by inducing HER2-IL13Rα2 heterodimers, which promoted superadditive T cell activation when both antigens were encountered concurrently. TanCAR T cell activity was more sustained but not more exhaustible than that of T cells that coexpressed a HER2 CAR and an IL13Rα2 CAR, T cells with a unispecific CAR, or a pooled product. In a murine glioblastoma model, TanCAR T cells mitigated antigen escape, displayed enhanced antitumor efficacy, and improved animal survival. Thus, TanCAR T cells show therapeutic potential to improve glioblastoma control by coengaging HER2 and IL13Rα2 in an augmented, bivalent immune synapse that enhances T cell functionality and reduces antigen escape.
Collapse
Affiliation(s)
- Meenakshi Hegde
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Malini Mukherjee
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Texas Children’s Hospital Center for Human Immunobiology, Houston, Texas, USA
| | - Zakaria Grada
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Antonella Pignata
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Daniel Landi
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Shoba A. Navai
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Amanda Wakefield
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Kristen Fousek
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Kevin Bielamowicz
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Kevin K.H. Chow
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Vita S. Brawley
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Tiara T. Byrd
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Simone Krebs
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Winfried S. Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Matthew L. Baker
- National Center for Macromolecular Imaging, Department of Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Gianpietro Dotti
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Malcolm K. Brenner
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
| | - Jordan S. Orange
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Texas Children’s Hospital Center for Human Immunobiology, Houston, Texas, USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Texas Children’s Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Cancer and Hematology Centers and
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics and
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
59
|
Müller I, Altherr D, Eyrich M, Flesch B, Friedmann KS, Ketter R, Oertel J, Schwarz EC, Technau A, Urbschat S, Eichler H. Tumor antigen-specific T cells for immune monitoring of dendritic cell-treated glioblastoma patients. Cytotherapy 2016; 18:1146-61. [PMID: 27424145 DOI: 10.1016/j.jcyt.2016.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AIMS CD8(+) T cells are part of the adaptive immune system and, as such, are responsible for the elimination of tumor cells. Dendritic cells (DC) are professional antigen-presenting cells (APC) that activate CD8(+) T cells. Effector CD8(+) T cells in turn mediate the active immunotherapeutic response of DC vaccination against the aggressive glioblastoma (GBM). The lack of tumor response assays complicates the assessment of treatment success in GBM patients. METHODS A novel assay to identify specific cytotoxicity of activated T cells by APC was evaluated. Tumor antigen-pulsed DCs from HLA-A*02-positive GBM patients were cultivated to stimulate autologous cytotoxic T lymphocytes (CTL) over a 12-day culture period. To directly correlate antigen specificity and cytotoxic capacity, intracellular interferon (IFN)-γ fluorescence flow cytometry-based measurements were combined with anti-GBM tumor peptide dextramer staining. IFN-γ response was quantified by real-time polymerase chain reaction (PCR), and selected GBM genes were compared with healthy human brain cDNA by single specific primer PCR characterization. RESULTS Using CTL of GBM patients stimulated with GBM lysate-pulsed DCs increased IFN-γ messenger RNA levels, and intracellular IFN-γ protein expression was positively correlated with specificity against GBM antigens. Moreover, the GBM peptide-specific CD8(+) T-cell response correlated with specific GBM gene expression. Following DC vaccination, GBM patients showed 10-fold higher tumor-specific signals compared with unvaccinated GBM patients. DISCUSSION These data indicate that GBM tumor peptide-dextramer staining of CTL in combination with intracellular IFN-γ staining may be a useful tool to acquire information on whether a specific tumor antigen has the potential to induce an immune response in vivo.
Collapse
Affiliation(s)
- Isabelle Müller
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Germany.
| | - Dominik Altherr
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Germany
| | - Matthias Eyrich
- Stem Cell Laboratory, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Brigitte Flesch
- Immungenetic/HLA, German Red Cross Blood Service, Bad Kreuznach, Germany
| | - Kim S Friedmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University School of Medicine, Homburg, Germany
| | - Ralf Ketter
- Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University School of Medicine, Homburg, Germany
| | - Antje Technau
- Stem Cell Laboratory, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Steffi Urbschat
- Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Hermann Eichler
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
60
|
Ladomersky E, Genet M, Zhai L, Gritsina G, Lauing KL, Lulla RR, Fangusaro J, Lenzen A, Kumthekar P, Raizer JJ, Binder DC, James CD, Wainwright DA. Improving vaccine efficacy against malignant glioma. Oncoimmunology 2016; 5:e1196311. [PMID: 27622066 DOI: 10.1080/2162402x.2016.1196311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 12/19/2022] Open
Abstract
The effective treatment of adult and pediatric malignant glioma is a significant clinical challenge. In adults, glioblastoma (GBM) accounts for the majority of malignant glioma diagnoses with a median survival of 14.6 mo. In children, malignant glioma accounts for 20% of primary CNS tumors with a median survival of less than 1 y. Here, we discuss vaccine treatment for children diagnosed with malignant glioma, through targeting EphA2, IL-13Rα2 and/or histone H3 K27M, while in adults, treatments with RINTEGA, Prophage Series G-100 and dendritic cells are explored. We conclude by proposing new strategies that are built on current vaccine technologies and improved upon with novel combinatorial approaches.
Collapse
Affiliation(s)
- Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Matthew Genet
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Galina Gritsina
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Rishi R Lulla
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Ann & Robert Lurie Children's Hospital of Northwestern University, Chicago, IL, USA
| | - Jason Fangusaro
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Ann & Robert Lurie Children's Hospital of Northwestern University, Chicago, IL, USA
| | - Alicia Lenzen
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Hematology, Oncology and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Ann & Robert Lurie Children's Hospital of Northwestern University, Chicago, IL, USA
| | - Priya Kumthekar
- Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jeffrey J Raizer
- Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David C Binder
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA; Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - C David James
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern Brain Tumor Institute, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
61
|
Ferguson SD, Srinivasan VM, Ghali MG, Heimberger AB. Cytomegalovirus-targeted immunotherapy and glioblastoma: hype or hope? Immunotherapy 2016; 8:413-23. [PMID: 26973123 DOI: 10.2217/imt.16.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Malignant gliomas, including glioblastoma (GBM), are the most common primary brain tumors. Despite extensive research only modest gains have been made in long-term survival. Standard of care involves maximizing safe surgical resection followed by concurrent chemoradiation with temozolomide. Immunotherapy for GBM is an area of intense research in recent years. New immunotherapies, although promising, have not been integrated into standard practice. Human cytomegalovirus (HCMV) is a DNA virus of the family Herpesviridae. Human seroprevalence is approximately 80%, and in most cases, is associated with asymptomatic infection. HCMV may be an important agent in the initiation, promotion and/or progression of tumorigenesis. Regardless of a possible etiologic role in GBM, interest has centered on exploiting this association for development of immunomodulatory therapies.
Collapse
Affiliation(s)
- Sherise D Ferguson
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Department of Neurosurgery, 1400 Holcombe Blvd, Unit 442, Houston, TX 77030, USA
| | - Visish M Srinivasan
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Department of Neurosurgery, 1400 Holcombe Blvd, Unit 442, Houston, TX 77030, USA
| | - Michael Gz Ghali
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, 2900 Queen Lane, PA, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Department of Neurosurgery, 1400 Holcombe Blvd, Unit 442, Houston, TX 77030, USA
| |
Collapse
|
62
|
Nakajima H, Murakami Y, Morii E, Akao T, Tatsumi N, Odajima S, Fukuda M, Machitani T, Iwai M, Kawata S, Hojo N, Oka Y, Sugiyama H, Oji Y. Induction of eEF2-specific antitumor CTL responses in vivo by vaccination with eEF2-derived 9mer-peptides. Oncol Rep 2016; 35:1959-66. [PMID: 26820500 DOI: 10.3892/or.2016.4589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/24/2015] [Indexed: 11/05/2022] Open
Abstract
Eukaryotic elongation factor 2 (eEF2) is an essential factor for protein synthesis. Previous studies have shown that the eEF2 gene was overexpressed and plays an oncogenic role in various types of cancers and that eEF2 gene product elicited both humoral immune responses to produce eEF2-specific IgG autoantibody in cancer-bearing individuals and cellular immune responses to induce eEF2 peptide-specific cytotoxic T lymphocytes (CTLs) in vitro. The purpose of the present study was to induce eEF2-specific, antitumor CTL responses in vivo by vaccination with MHC class I-binding eEF2-derived peptide. First, two mouse MHC class I-restricted eEF2‑derived, 9-mer peptides, EF17 (17-25 aa, ANIRNMSVI) and EF180 (180-188 aa, RIVENVNVI) were identified as eEF2-specific CTL peptides, and mice were vaccinated intradermally eight times with either EF17 or EF180 peptide emulsified with Montanide ISA51 adjuvant. Cytotoxicity assay showed that eEF2-specific CTLs were induced in both EF17‑and EF180‑vaccinated mice, and histological study showed no detectable damage in the organs of these mice. Next, to examine in vivo antitumor effects of eEF2 peptide vaccination in a therapeutic model, mice were vaccinated four times with one each of the two eEF2 peptides at weekly intervals after implantation of eEF2-expressing leukemia cells. The vaccination with eEF2 peptides induced eEF2-specific CTLs and suppressed tumor growth, and disease-free survival was significantly longer in EF180-vaccinated mice compared to control mice. The survival was associated with the robustness of eEF2-specific CTL induction. These results indicate that vaccination with MHC class I-binding eEF2 peptide induced eEF2-targeting, antitumor CTL responses in vivo without damage to normal organs, which provided us a rationale for eEF2 peptide-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yui Murakami
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshiki Akao
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoya Tatsumi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoko Odajima
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mari Fukuda
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takao Machitani
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Miki Iwai
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sayo Kawata
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nozomi Hojo
- Department of Functional Diagnostic Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Oji
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
63
|
Abstract
Gliomas are the most common primary brain tumors of the central nervous system, and carry a grim prognosis. Novel approaches utilizing the immune system as adjuvant therapy are quickly emerging as viable and effective options. Immunotherapeutic strategies being investigated to treat glioblastoma include: vaccination therapy targeted against either specific tumor antigens or whole tumor lysate, adoptive cellular therapy with cytotoxic T lymphocytes, chimeric antigen receptors and bi-specific T-cell engaging antibodies allowing circumvention of major histocompatibility complex restriction, aptamer therapy with aims for more efficient target delivery, and checkpoint blockade in order to release the tumor-mediated inhibition of the immune system. Given the heterogeneity of glioblastoma and its ability to gain mutations throughout the disease course, multifaceted treatment strategies utilizing multiple forms of immunotherapy in combination with conventional therapy will be most likely to succeed moving forward.
Collapse
Affiliation(s)
- Brandon D Liebelt
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Houston Methodist Neurological Institute, Houston, TX, USA
| | - Gaetano Finocchiaro
- Department of Neuro-oncology, IRCCS Istituto Neurologico Besta, Milan, Italy
| | - Amy B Heimberger
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
64
|
Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, Hattingen E, Harter PN, Mittelbronn M, Tonn T, Steinbach JP, Wels WS. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J Natl Cancer Inst 2015; 108:djv375. [PMID: 26640245 DOI: 10.1093/jnci/djv375] [Citation(s) in RCA: 261] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/02/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and malignant intracranial tumor in adults and currently incurable. To specifically target natural killer (NK) cell activity to GBM, we employed NK-92/5.28.z cells that are continuously expanding human NK cells expressing an ErbB2-specific chimeric antigen receptor (CAR). METHODS ErbB2 expression in 56 primary tumors, four primary cell cultures, and seven established cell lines was assessed by immunohistochemistry and flow cytometry. Cell killing activity of NK-92/5.28.z cells was analyzed in in vitro cytotoxicity assays. In vivo antitumor activity was evaluated in NOD-SCID IL2Rγ(null) (NSG) mice carrying orthotopic human GBM xenografts (6 to 11 mice per group) and C57BL/6 mice carrying subcutaneous and orthotopic ErbB2-expressing murine GBM tumors (5 to 8 mice per group). Statistical tests were two-sided. RESULTS We found elevated ErbB2 protein expression in 41% of primary GBM samples and in the majority of GBM cell lines investigated. In in vitro assays, NK-92/5.28.z in contrast to untargeted NK-92 cells lysed all ErbB2-positive established and primary GBM cells analyzed. Potent in vivo antitumor activity of NK-92/5.28.z was observed in orthotopic GBM xenograft models in NSG mice, leading to a marked extension of symptom-free survival upon repeated stereotactic injection of CAR NK cells into the tumor area (median survival of 200.5 days upon treatment with NK-92/5.28.z vs 73 days upon treatment with parental NK-92 cells, P < .001). In immunocompetent mice, local therapy with NK-92/5.28.z cells resulted in cures of transplanted syngeneic GBM in four of five mice carrying subcutaneous tumors and five of eight mice carrying intracranial tumors, induction of endogenous antitumor immunity, and long-term protection against tumor rechallenge at distant sites. CONCLUSIONS Our data demonstrate the potential of ErbB2-specific NK-92/5.28.z cells for adoptive immunotherapy of glioblastoma, justifying evaluation of this approach for the treatment of ErbB2-positive GBM in clinical studies.
Collapse
Affiliation(s)
- Congcong Zhang
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Michael C Burger
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Lukas Jennewein
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Sabrina Genßler
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Kurt Schönfeld
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Pia Zeiner
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Elke Hattingen
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Patrick N Harter
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Michel Mittelbronn
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Torsten Tonn
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Joachim P Steinbach
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Winfried S Wels
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| |
Collapse
|
65
|
Is CMV a target in pediatric glioblastoma? Expression of CMV proteins, pp65 and IE1-72 and CMV nucleic acids in a cohort of pediatric glioblastoma patients. J Neurooncol 2015; 125:307-15. [PMID: 26341370 PMCID: PMC4612329 DOI: 10.1007/s11060-015-1905-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 08/29/2015] [Indexed: 12/30/2022]
Abstract
While the 5-year overall survival is better in pediatric than in adult patients diagnosed with glioblastoma (GBM), outcomes in children remain very poor. Understanding the mechanisms of tumorigenesis and tumor propagation can identify therapeutic targets to improve these outcomes. Human cytomegalovirus (CMV) proteins and nucleic acids are present in the majority of adult GBM. Indeed, CMV is emerging as a potential glioma-associated target for anti-CMV agents and cellular therapeutics. Furthermore, CMV appears to contribute to GBM’s malignant phenotype, although its role in tumorigenesis is less certain. In this cohort of 25 serially diagnosed pediatric GBMs, the largest described cohort to date, we used immunohistochemical staining and in situ hybridization to show the presence of CMV antigens pp65 and IE1-72 as well as CMV nucleic acids, respectively. Our cohort indicated either CMV antigen pp65 or IE1-72 was present in approximately 67 % of pediatric GBM samples. The majority of samples stained positive for either CMV antigen showing a cytoplasmic pattern in 25-50 % of cells within the sample at a moderate intensity, while a few samples showed nuclear staining and higher grade/intensity. Of 16 samples where in situ hybridization was performed, 13 (81 %) showed specific staining using a CMV genome specific probe cocktail. ISH positive samples showed high concordance with being pp65 or IE1-72 positive. These findings, paired with the association of CMV expression with poor prognosis and overall survival, indicate the need to further investigate how these antigens are promoting tumor growth and preventing cell death. Also, the expression of these antigens in a majority of tumor tissues should be considered for immunotherapeutic targets in cases of pediatric GBM.
Collapse
|
66
|
Kang JH, Adamson C. Novel chemotherapeutics and other therapies for treating high-grade glioma. Expert Opin Investig Drugs 2015; 24:1361-79. [PMID: 26289791 DOI: 10.1517/13543784.2015.1048332] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Despite extensive research, high-grade glioma (HGG) remains a dire diagnosis with no change in the standard of care in almost a decade. However, recent advancements uncovering molecular biomarkers of brain tumors and tumor-specific antigens targeted by immunotherapies provide opportunities for novel personalized treatment regimens to improve survival. AREAS COVERED In this review, the authors provide a comprehensive overview of recent therapeutic advancements in HGG. Furthermore, they describe new molecular biomarkers and molecular classifications, in addition to updated research on bevacizumab, targeted molecular therapies, immunotherapy and alternative delivery methods that overcome the blood-brain barrier to reach the target tumor tissue. Challenges regarding each therapy are also outlined. The authors also provide some insight into a novel non-chemotherapeutic treatment for malignant glioma, NovoTTFA, as well as a summary of current treatment options for recurrence. EXPERT OPINION Current research for treating malignant gliomas are paving the path to personalized therapy, including immunotherapy, that involve integrated genomic and histolopathologic data, as well as a multi-modal treatment regimen. Immunotherapy will potentially be the next addition to the current standard of care, specialized to the antigens presented on the tumors. The results of the current trials of multi-antigen vaccines are eagerly anticipated.
Collapse
Affiliation(s)
- Jennifer H Kang
- a 1 Duke University School of Medicine , Box 3807, Durham, NC, USA
| | - Cory Adamson
- b 2 Director, Molecular Neuro-oncology Lab, Duke Medical Center , DUMC Box 3807, Durham, NC, USA.,c 3 Chief of Neurosurgery, Durham VA Medical Center , 508 Fulton Street, Durham, NC, USA +1 919 698 3152 ; .,d 4 Duke Medical Center , DUMC Box 3807, Durham, NC, USA
| |
Collapse
|
67
|
Dendritic Cell-Based Immunotherapy Treatment for Glioblastoma Multiforme. BIOMED RESEARCH INTERNATIONAL 2015; 2015:717530. [PMID: 26167495 PMCID: PMC4488155 DOI: 10.1155/2015/717530] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/08/2015] [Indexed: 12/23/2022]
Abstract
Glioblastoma multiforme (GBM) is the most malignant glioma and patients diagnosed with this disease had poor outcomes even treated with the combination of conventional treatment (surgery, chemotherapy, and radiation). Dendritic cells (DCs) are the most powerful antigen presenting cells and DC-based vaccination has the potential to target and eliminate GBM cells and enhance the responses of these cells to the existing therapies with minimal damage to the healthy tissues around them. It can enhance recognition of GBM cells by the patients' immune system and activate vast, potent, and long-lasting immune reactions to eliminate them. Therefore, this therapy can prolong the survival of GBM patients and has wide and bright future in the treatment of GBM. Also, the efficacy of this therapy can be strengthened in several ways at some degree: the manipulation of immune regulatory components or costimulatory molecules on DCs; the appropriate choices of antigens for loading to enhance the effectiveness of the therapy; regulation of positive regulators or negative regulators in GBM microenvironment.
Collapse
|
68
|
Combining immunotherapy with radiation for the treatment of glioblastoma. J Neurooncol 2015; 123:459-64. [PMID: 25877468 DOI: 10.1007/s11060-015-1762-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/28/2015] [Indexed: 01/09/2023]
Abstract
Glioblastoma is a devastating cancer with universally poor outcomes in spite of current standard multimodal therapy. Immunotherapy is an attractive new treatment modality given its potential for exquisite specificity and its favorable side effect profile; however, clinical trials of immunotherapy in GBM have thus far shown modest benefit. Optimally combining radiation with immunotherapy may be the key to unlocking the potential of both therapies given the evidence that radiation can enhance anti-tumor immunity. Here we review this evidence and discuss considerations for combined therapy.
Collapse
|
69
|
Biomarkers for glioma immunotherapy: the next generation. J Neurooncol 2015; 123:359-72. [PMID: 25724916 DOI: 10.1007/s11060-015-1746-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/16/2015] [Indexed: 12/11/2022]
Abstract
The term "biomarker" historically refers to a single parameter, such as the expression level of a gene or a radiographic pattern, used to indicate a broader biological state. Molecular indicators have been applied to several aspects of cancer therapy: to describe the genotypic and phenotypic state of neoplastic tissue for prognosis, to predict susceptibility to anti-proliferative agents, to validate the presence of specific drug targets, and to evaluate responsiveness to therapy. For glioblastoma (GBM), immunohistochemical and radiographic biomarkers accessible to the clinical lab have informed traditional regimens, but while immunotherapies have emerged as potentially disruptive weapons against this diffusely infiltrating, heterogeneous tumor, biomarkers with strong predictive power have not been fully established. The cancer immunotherapy field, through the recently accelerated expansion of trials, is currently leveraging this wealth of clinical and biological data to define and revise the use of biomarkers for improving prognostic accuracy, personalization of therapy, and evaluation of responses across the wide variety of tumors. Technological advancements in DNA sequencing, cytometry, and microscopy have facilitated the exploration of more integrated, high-dimensional profiling of the disease system-incorporating both immune and tumor parameters-rather than single metrics, as biomarkers for therapeutic sensitivity. Here we discuss the utility of traditional GBM biomarkers in immunotherapy and how the impending transformation of the biomarker paradigm-from single markers to integrated profiles-may offer the key to bringing predictive, personalized immunotherapy to GBM patients.
Collapse
|
70
|
Li X, Mei Q, Nie J, Fu X, Han W. Decitabine: a promising epi-immunotherapeutic agent in solid tumors. Expert Rev Clin Immunol 2015; 11:363-75. [DOI: 10.1586/1744666x.2015.1002397] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
71
|
Emerging Strategies for the Treatment of Tumor Stem Cells in Central Nervous System Malignancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:167-87. [DOI: 10.1007/978-3-319-16537-0_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
72
|
Calinescu AA, Kamran N, Baker G, Mineharu Y, Lowenstein PR, Castro MG. Overview of current immunotherapeutic strategies for glioma. Immunotherapy 2015; 7:1073-104. [PMID: 26598957 PMCID: PMC4681396 DOI: 10.2217/imt.15.75] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last decade, numerous studies of immunotherapy for malignant glioma (glioblastoma multiforme) have brought new knowledge and new hope for improving the prognosis of this incurable disease. Some clinical trials have reached Phase III, following positive outcomes in Phase I and II, with respect to safety and immunological end points. Results are encouraging especially when considering the promise of sustained efficacy by inducing antitumor immunological memory. Progress in understanding the mechanisms of tumor-induced immune suppression led to the development of drugs targeting immunosuppressive checkpoints, which are used in active clinical trials for glioblastoma multiforme. Insights related to the heterogeneity of the disease bring new challenges for the management of glioma and underscore a likely cause of therapeutic failure. An emerging therapeutic strategy is represented by a combinatorial, personalized approach, including the standard of care: surgery, radiation, chemotherapy with added active immunotherapy and multiagent targeting of immunosuppressive checkpoints.
Collapse
Affiliation(s)
| | - Neha Kamran
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Gregory Baker
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University, Kyoto, Japan
| | - Pedro Ricardo Lowenstein
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria Graciela Castro
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
73
|
Reardon DA, Freeman G, Wu C, Chiocca EA, Wucherpfennig KW, Wen PY, Fritsch EF, Curry WT, Sampson JH, Dranoff G. Immunotherapy advances for glioblastoma. Neuro Oncol 2014; 16:1441-58. [PMID: 25190673 DOI: 10.1093/neuonc/nou212] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Survival for patients with glioblastoma, the most common high-grade primary CNS tumor, remains poor despite multiple therapeutic interventions including intensifying cytotoxic therapy, targeting dysregulated cell signaling pathways, and blocking angiogenesis. Exciting, durable clinical benefits have recently been demonstrated for a number of other challenging cancers using a variety of immunotherapeutic approaches. Much modern research confirms that the CNS is immunoactive rather than immunoprivileged. Preliminary results of clinical studies demonstrate that varied vaccine strategies have achieved encouraging evidence of clinical benefit for glioblastoma patients, although multiple variables will likely require systematic investigation before optimal outcomes are realized. Initial preclinical studies have also revealed promising results with other immunotherapies including cell-based approaches and immune checkpoint blockade. Clinical studies to evaluate a wide array of immune therapies for malignant glioma patients are being rapidly developed. Important considerations going forward include optimizing response assessment and identifiying correlative biomarkers for predict therapeutic benefit. Finally, the potential of complementary combinatorial immunotherapeutic regimens is highly exciting and warrants expedited investigation.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Gordon Freeman
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Catherine Wu
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - E Antonio Chiocca
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Kai W Wucherpfennig
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Edward F Fritsch
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - William T Curry
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - John H Sampson
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Glenn Dranoff
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| |
Collapse
|
74
|
Lanitis E, Smith JB, Dangaj D, Flingai S, Poussin M, Xu S, Czerniecki BJ, Li YF, Robbins PF, Powell DJ. A human ErbB2-specific T-cell receptor confers potent antitumor effector functions in genetically engineered primary cytotoxic lymphocytes. Hum Gene Ther 2014; 25:730-9. [PMID: 25003657 PMCID: PMC4137348 DOI: 10.1089/hum.2014.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/20/2014] [Indexed: 01/10/2023] Open
Abstract
The ErbB2 protein is a member of the tyrosine kinase family of growth factor receptors that is overexpressed in cancers of the breast, ovary, stomach, kidney, colon, and lung, and therefore represents an attractive candidate antigen for targeted cancer immunotherapy. Cytotoxic T lymphocytes specific for various immunogenic ErbB2 peptides have been described, but they often exhibit both poor functional avidity and tumor reactivity. In order to generate potent CD8(+) T cells with specificity for the ErbB2(369-377) peptide, we performed one round of in vitro peptide stimulation of CD8(+) T cells isolated from an HLA-A2(+) patient who was previously vaccinated with autologous dendritic cells pulsed with HLA class I ErbB2 peptides. Using this approach, we enriched highly avid ErbB2-reactive T cells with strong ErbB2-specific, antitumor effector functions. We then stimulated these ErbB2-reactive T cells with ErbB2(+) HLA-A2(+) tumor cells in vitro and sorted tumor-activated ErbB2(369-377) peptide T cells, which allowed for the isolation of a novel T-cell receptor (TCR) with ErbB2(369-377) peptide specificity. Primary human CD8(+) T cells genetically modified to express this ErbB2-specific TCR specifically bound ErbB2(369-377) peptide containing HLA-A2 tetramers, and efficiently recognized target cells pulsed with low nanomolar concentrations of ErbB2(369-377) peptide as well as nonpulsed ErbB2(+) HLA-A2(+) tumor cell lines in vitro. In a novel xenograft model, ErbB2-redirected T cells also significantly delayed progression of ErbB2(+) HLA-A2(+) human tumor in vivo. Together, these results support the notion that redirection of normal T-cell specificity by TCR gene transfer can have potential applications in the adoptive immunotherapy of ErbB2-expressing malignancies.
Collapse
Affiliation(s)
- Evripidis Lanitis
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Jenessa B. Smith
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Denarda Dangaj
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Seleeke Flingai
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Mathilde Poussin
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104
| | - Brian J. Czerniecki
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia, PA 19104
| | - Yong F. Li
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Paul F. Robbins
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Daniel J. Powell
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104
- Abramson Cancer Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
75
|
Glioma-associated antigen HEATR1 induces functional cytotoxic T lymphocytes in patients with glioma. J Immunol Res 2014; 2014:131494. [PMID: 25126583 PMCID: PMC4121097 DOI: 10.1155/2014/131494] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/17/2014] [Accepted: 06/16/2014] [Indexed: 11/18/2022] Open
Abstract
A2B5+ glioblastoma (GBM) cells have glioma stem-like cell (GSC) properties that are crucial to chemotherapy resistance and GBM relapse. T-cell-based antigens derived from A2B5+ GBM cells provide important information for immunotherapy. Here, we show that HEAT repeat containing 1 (HEATR1) expression in GBM tissues was significantly higher than that in control brain tissues. Furthermore, HEATR1 expression in A2B5+ U87 cells was higher than that in A2B5-U87 cells (P = 0.016). Six peptides of HEATR1 presented by HLA-A∗02 were selected for testing of their ability to induce T-cell responses in patients with GBM. When peripheral blood mononuclear cells from healthy donors (n = 6) and patients with glioma (n = 33) were stimulated with the peptide mixture, eight patients with malignant gliomas had positive reactivity with a significantly increased number of responding T-cells. The peptides HEATR(1682-690), HEATR(11126-1134), and HEATR(1757-765) had high affinity for binding to HLA-A∗02:01 and a strong capacity to induce CTL response. CTLs against HEATR1 peptides were capable of recognizing and lysing GBM cells and GSCs. These data are the first to demonstrate that HEATR1 could induce specific CTL responses targeting both GBM cells and GSCs, implicating that HEATR1 peptide-based immunotherapy could be a novel promising strategy for treating patients with GBM.
Collapse
|
76
|
A new hope in immunotherapy for malignant gliomas: adoptive T cell transfer therapy. J Immunol Res 2014; 2014:326545. [PMID: 25009822 PMCID: PMC4070364 DOI: 10.1155/2014/326545] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/02/2014] [Accepted: 05/18/2014] [Indexed: 11/18/2022] Open
Abstract
Immunotherapy emerged as a promising therapeutic approach to highly incurable malignant gliomas due to tumor-specific cytotoxicity, minimal side effect, and a durable antitumor effect by memory T cells. But, antitumor activities of endogenously activated T cells induced by immunotherapy such as vaccination are not sufficient to control tumors because tumor-specific antigens may be self-antigens and tumors have immune evasion mechanisms to avoid immune surveillance system of host. Although recent clinical results from vaccine strategy for malignant gliomas are encouraging, these trials have some limitations, particularly their failure to expand tumor antigen-specific T cells reproducibly and effectively. An alternative strategy to overcome these limitations is adoptive T cell transfer therapy, in which tumor-specific T cells are expanded ex vivo rapidly and then transferred to patients. Moreover, enhanced biologic functions of T cells generated by genetic engineering and modified immunosuppressive microenvironment of host by homeostatic T cell expansion and/or elimination of immunosuppressive cells and molecules can induce more potent antitumor T cell responses and make this strategy hold promise in promoting a patient response for malignant glioma treatment. Here we will review the past and current progresses and discuss a new hope in adoptive T cell therapy for malignant gliomas.
Collapse
|
77
|
Xu LW, Chow KKH, Lim M, Li G. Current vaccine trials in glioblastoma: a review. J Immunol Res 2014; 2014:796856. [PMID: 24804271 PMCID: PMC3996322 DOI: 10.1155/2014/796856] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/12/2014] [Accepted: 02/28/2014] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor, and despite aggressive therapy with surgery, radiation, and chemotherapy, average survival remains at about 1.5 years. The highly infiltrative and invasive nature of GBM requires that alternative treatments for this disease be widespread and targeted to tumor cells. Immunotherapy in the form of tumor vaccines has the potential to meet this need. Vaccines against GBM hold the promise of triggering specific and systemic antitumor immune responses that may be the key to eradicating this unrelenting cancer. In this review, we will discuss past and present clinical trials of various GBM vaccines and their potential impact on the future care of GBM patients. There have been many promising phase I and phase II GBM vaccine studies that have led to ongoing and upcoming phase III trials. If the results of these randomized trials show a survival benefit, immunotherapy will become a standard part of the treatment of this devastating disease.
Collapse
Affiliation(s)
- Linda W. Xu
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| | - Kevin K. H. Chow
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Gordon Li
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| |
Collapse
|
78
|
Akiyama Y, Komiyama M, Miyata H, Yagoto M, Ashizawa T, Iizuka A, Oshita C, Kume A, Nogami M, Ito I, Watanabe R, Sugino T, Mitsuya K, Hayashi N, Nakasu Y, Yamaguchi K. Novel cancer-testis antigen expression on glioma cell lines derived from high-grade glioma patients. Oncol Rep 2014; 31:1683-90. [PMID: 24573400 DOI: 10.3892/or.2014.3049] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/29/2014] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant and aggressive tumors, and has a very poor prognosis with a mean survival time of <2 years, despite intensive treatment using chemo-radiation. Therefore, novel therapeutic approaches including immunotherapy have been developed against GBM. For the purpose of identifying novel target antigens contributing to GBM treatment, we developed 17 primary glioma cell lines derived from high-grade glioma patients, and analyzed the expression of various tumor antigens and glioma-associated markers using a quantitative PCR and immunohistochemistry (IHC). A quantitative PCR using 54 cancer-testis (CT) antigen-specific primers showed that 36 CT antigens were positive in at least 1 of 17 serum-derived cell lines, and 17 antigens were positive in >50% cell lines. Impressively, 6 genes (BAGE, MAGE-A12, CASC5, CTAGE1, DDX43 and IL-13RA2) were detected in all cell lines. The expression of other 13 glioma-associated antigens than CT genes were also investigated, and 10 genes were detected in >70% cell lines. The expression of CT antigen and glioma-associated antigen genes with a high frequency were also verified in IHC analysis. Moreover, a relationship of antigen gene expressions with a high frequency to overall survival was investigated using the Repository of Molecular Brain Neoplasia Data (REMBRANDT) database of the National Cancer Institute, and expression of 6 genes including IL-13RA2 was inversely correlated to overall survival time. Furthermore, 4 genes including DDX43, TDRD1, HER2 and gp100 were identified as MGMT-relevant factors. In the present study, several CT antigen including novel genes were detected in high-grade glioma primary cell lines, which might contribute to developing novel immunotherapy and glioma-specific biomarkers in future.
Collapse
Affiliation(s)
- Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Masaru Komiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Mika Yagoto
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Akira Iizuka
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Chie Oshita
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Akiko Kume
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Masahiro Nogami
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ichiro Ito
- Division of Pathology, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka 411-8777, Japan
| | - Reiko Watanabe
- Division of Pathology, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka 411-8777, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka 411-8777, Japan
| | - Koichi Mitsuya
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka 411-8777, Japan
| | - Nakamasa Hayashi
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yoko Nakasu
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka 411-8777, Japan
| |
Collapse
|
79
|
Abstract
Despite dramatic advances in surgical techniques, imaging and adjuvant radiotherapy or chemotherapy, the prognosis for patients with malignant glial tumors remains dismal. Based on the current knowledge regarding immune responses in the healthy CNS and glioma-bearing hosts, this review discusses dendritic cell-based immunotherapeutic approaches for malignant gliomas and the relevance of recent clinical trials and their outcomes. It is now recognized that the CNS is not an immunologically tolerated site and clearance of arising glioma cells is a routine physiologic function of the normal, noncompromised immune system. To escape from immune surveillance, however, clinically apparent gliomas develop complex mechanisms that suppress tumoricidal immune responses. Although the use of dendritic cells for the treatment of glioma patients may be the most appropriate approach, an effective treatment paradigm for these tumors may eventually require the use of several types of treatment. Additionally, given the heterogeneity of this disease process and an immune-refractory tumor cell population, the series use of rational multiple modalities that target disparate tumor characteristics may be the most effective therapeutic strategy to treat malignant gliomas.
Collapse
Affiliation(s)
- Yasuharu Akasaki
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Suite 800 East, 8631 West 3 Street, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
80
|
Olin MR, Pluhar GE, Andersen BM, Shaver R, Waldron NN, Moertel CL. Victory and defeat in the induction of a therapeutic response through vaccine therapy for human and canine brain tumors: a review of the state of the art. Crit Rev Immunol 2014; 34:399-432. [PMID: 25404047 PMCID: PMC4485925 DOI: 10.1615/critrevimmunol.2014011577] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Anti-tumor immunotherapy using tumor lysate-based vaccines has made great advances over recent decades. Cancer vaccines aim to elicit adaptive immune responses through various pathways by providing tumor and tumor-associated antigens with an immune stimulant or adjuvant. These anti-tumor vaccines are therefore developed as personalized treatments. Utilizing tumors as a source of vaccine antigens in immunotherapy has demonstrated promising results with minimal toxicity. However, to date, researchers have failed to overcome the overpowering immune suppressive effects within the tumor microenvironment. Immune suppression occurs naturally via multiple mechanisms. These mechanisms serve an important homeostatic role restoring a normal tissue microenvironment following an inflammatory response. Due to these suppressive mechanisms and the inherent heterogeneity of tumors, it is imperative to then elicit and maintain a specific tumoricidal response if vaccine therapy or some other combination of reagents is chosen. In this review, we focus on the historical use of tumors as a source of antigens to elicit a tumoricidal response and the limitations encountered that prevent greater success in immunotherapy. We describe the advantages and disadvantages of various vaccines and their ineffectiveness due to tumor-induced immune suppression.
Collapse
Affiliation(s)
- Michael R. Olin
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - G. Elizabeth Pluhar
- Department of Veterinary Medicine, College of Veterinary Medicine. University of Minnesota, St. Paul, MN 55108
| | - Brian M. Andersen
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - Rob Shaver
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - Nate N. Waldron
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | | |
Collapse
|
81
|
Reardon DA, Wucherpfennig KW, Freeman G, Wu CJ, Chiocca EA, Wen PY, Curry WT, Mitchell DA, Fecci PE, Sampson JH, Dranoff G. An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev Vaccines 2013; 12:597-615. [PMID: 23750791 DOI: 10.1586/erv.13.41] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Outcome for glioblastoma (GBM), the most common primary CNS malignancy, remains poor. The overall survival benefit recently achieved with immunotherapeutics for melanoma and prostate cancer support evaluation of immunotherapies for other challenging cancers, including GBM. Much historical dogma depicting the CNS as immunoprivileged has been replaced by data demonstrating CNS immunocompetence and active interaction with the peripheral immune system. Several glioma antigens have been identified for potential immunotherapeutic exploitation. Active immunotherapy studies for GBM, supported by preclinical data, have focused on tumor lysate and synthetic antigen vaccination strategies. Results to date confirm consistent safety, including a lack of autoimmune reactivity; however, modest efficacy and variable immunogenicity have been observed. These findings underscore the need to optimize vaccination variables and to address challenges posed by systemic and local immunosuppression inherent to GBM tumors. Additional immunotherapy strategies are also in development for GBM. Future studies may consider combinatorial immunotherapy strategies with complimentary actions.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Mullins CS, Walter A, Schmitt M, Classen CF, Linnebacher M. Tumor antigen and MHC expression in glioma cells for immunotherapeutic interventions. World J Immunol 2013; 3:62-67. [DOI: 10.5411/wji.v3.i3.62] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/15/2013] [Accepted: 11/03/2013] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate the expression of tumor-antigens and major histocompatibility complex (MHC)-machinery components in glioblastoma multiforme cell lines flow cytometry staining methods were applied.
METHODS: Ten GBM cell lines (three commercially available: U-87 MG, U-138-MG and GMS-10 as well as seven newly established cell lines from individual patients in low-passages: HROG02, HROG04, HROG05, HROG06, HROG10, HROG13 and HROG17) were analyzed for expression of (I) general and (II) GBM-related tumor antigens as well as of (III) components of the MHC machinery by flow cytometry.
RESULTS: All cell lines expressed MHC class I with seven out of the ten being HLA-A02 positive. Four of the seven primary cell lines additionally expressed MHC class II in a constitutive manner. Of note, after interferon gamma (IFN-γ) treatment, all seven cell lines expressed MHC class II. The tumor associated antigens (TAA) EGFR and survivin were expressed at high levels in all cell lines; whereas MART-1, RHAMM, WT-1 and IL-13Rα were expressed by at least half of the cell lines and HER2/neu, MAGE-1 and tyrosinase were expressed only by few cell lines. However, all cell lines expressed at least two of the candidate antigens included into this analysis.
CONCLUSION: No obvious differences between commercially available and newly-established cell lines were observed. Thus, the latter in low-passages are interesting for (therapy-) screening and immunotherapeutic strategies.
Collapse
|
83
|
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: 3.6] [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
|
84
|
Hegde M, Corder A, Chow KKH, Mukherjee M, Ashoori A, Kew Y, Zhang YJ, Baskin DS, Merchant FA, Brawley VS, Byrd TT, Krebs S, Wu MF, Liu H, Heslop HE, Gottschalk S, Gottachalk S, Yvon E, Ahmed N. Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma. Mol Ther 2013; 21:2087-101. [PMID: 23939024 PMCID: PMC3831041 DOI: 10.1038/mt.2013.185] [Citation(s) in RCA: 279] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 07/30/2013] [Indexed: 12/18/2022] Open
Abstract
Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER2, IL-13Rα2, and EphA2 in primary GBM samples using multicolor flow cytometry and immunofluorescence, and applied a binomial routine to the permutations of antigen expression and the related odds of complete tumor elimination. This mathematical model demonstrated that cotargeting HER2 and IL-13Rα2 could maximally expand the therapeutic reach of the T cell product in all primary tumors studied. Targeting a third antigen did not predict an added advantage in the tumor cohort studied. We therefore generated bispecific T cell products from healthy donors and from GBM patients by pooling T cells individually expressing HER2 and IL-13Rα2-specific CARs and by making individual T cells to coexpress both molecules. Both HER2/IL-13Rα2-bispecific T cell products offset antigen escape, producing enhanced effector activity in vitro immunoassays (against autologous glioma cells in the case of GBM patient products) and in an orthotopic xenogeneic murine model. Further, T cells coexpressing HER2 and IL-13Rα2-CARs exhibited accentuated yet antigen-dependent downstream signaling and a particularly enhanced antitumor activity.
Collapse
Affiliation(s)
- Meenakshi Hegde
- 1] Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA [2] Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA [3] Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Khan-Farooqi HR, Prins RM, Liau LM. Tumor immunology, immunomics and targeted immunotherapy for central nervous system malignancies. Neurol Res 2013; 27:692-702. [PMID: 16197806 DOI: 10.1179/016164105x49490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Although the brain was traditionally considered as 'immunologically privileged', recent findings have implied an involvement of immune mechanisms in neurological disease and illness, including central nervous system (CNS) malignancies. In this review, we initially focus on aspects of the immune system critical for effective antitumor immunity, as an understanding of normal immunological functions and how they relate to tumor immunology will set a foundation for understanding the unique challenges facing the integration of neuro-oncology and neuroimmunology. We summarize current knowledge of immune responses in the 'immunologically quiescent' brain and its role in tumor immunology. We will then discuss the emerging field of 'immunomics' and recent advances in molecular technologies, such as DNA microarray, which are being applied to brain tumor antigen epitope discovery and patient stratification for brain cancer immunotherapy. This, in turn, should have significant importance for ultimately designing and developing efficient and focused strategies for anticancer immunotherapy. Finally, the current state of immune-based treatment paradigms and future directions will be discussed, paying particular attention to targeted antibody strategies, adoptive cellular immunotherapy, and tumor vaccine approaches that have been studied in clinical trials for CNS neoplasms.
Collapse
Affiliation(s)
- Haumith R Khan-Farooqi
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California 90095-6901, USA
| | | | | |
Collapse
|
86
|
Carén H, Pollard SM, Beck S. The good, the bad and the ugly: epigenetic mechanisms in glioblastoma. Mol Aspects Med 2013; 34:849-62. [PMID: 22771539 PMCID: PMC3714597 DOI: 10.1016/j.mam.2012.06.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 06/27/2012] [Indexed: 12/31/2022]
Abstract
Cell type-specific patterns of gene expression reflect epigenetic changes imposed through a particular developmental lineage as well as those triggered by environmental cues within adult tissues. There is great interest in elucidating the molecular basis and functional importance of epigenetic mechanisms in both normal physiology and disease - particularly in cancer, where abnormal '-omic' states are often observed. In this article we review recent progress in studies of epigenetic mechanisms in the most common primary adult brain cancer, glioblastoma multiforme. Three distinct areas are discussed. First, the evidence in support of ongoing 'normal' epigenetic processes associated with differentiation - as predicted by 'cancer stem cell' models of the disease. Second, identification of site-specific and global epigenetic abnormalities. Third, genetic disruptions directly within the core epigenetic machinery, exemplified by the recently identified mutations within isocitrate dehydrogenase genes IDH1/2 and variant histone genes H3.3/H3F3A. These constitute the 'good, the bad and the ugly' of epigenetic mechanisms in cancer.
Collapse
Affiliation(s)
- Helena Carén
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom
| | - Steven M. Pollard
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom
| | - Stephan Beck
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom
| |
Collapse
|
87
|
Glioma stem cells and immunotherapy for the treatment of malignant gliomas. ISRN ONCOLOGY 2013; 2013:673793. [PMID: 23762610 PMCID: PMC3671309 DOI: 10.1155/2013/673793] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 03/27/2013] [Indexed: 02/06/2023]
Abstract
Stem cell research has led to the discovery of glioma stem cells (GSCs), and because these cells are resistant to chemotherapy and radiotherapy, analysis of their properties has been rapidly pursued for targeted treatment of malignant glioma. Recent studies have also revealed complex crosstalk between GSCs and their specialized environment (niche). Therefore, targeting not only GSCs but also their niche may be a principle for novel therapies of malignant glioma. One possible novel strategy for targeting GSCs and their niches is immunotherapy with different antitumor mechanism(s) from those of conventional therapy. Recent clinical studies of immunotherapy using peptide vaccines and antibodies have shown promising results. This review describes the recent findings related to GSCs and their niches, as well as immunotherapies for glioma, followed by discussion of immunotherapies that target GSCs for the treatment of malignant glioma.
Collapse
|
88
|
Dunn GP, Fecci PE, Curry WT. Cancer immunoediting in malignant glioma. Neurosurgery 2013; 71:201-22; discussion 222-3. [PMID: 22353795 DOI: 10.1227/neu.0b013e31824f840d] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Significant work from many laboratories over the last decade in the study of cancer immunology has resulted in the development of the cancer immunoediting hypothesis. This contemporary framework of the naturally arising immune system-tumor interaction is thought to comprise 3 phases: elimination, wherein immunity subserves an extrinsic tumor suppressor function and destroys nascent tumor cells; equilibrium, wherein tumor cells are constrained in a period of latency under immune control; and escape, wherein tumor cells outpace immunity and progress clinically. In this review, we address in detail the relevance of the cancer immunoediting concept to neurosurgeons and neuro-oncologists treating and studying malignant glioma by exploring the de novo immune response to these tumors, how these tumors may persist in vivo, the mechanisms by which these cells may escape/attenuate immunity, and ultimately how this concept may influence our immunotherapeutic approaches.
Collapse
Affiliation(s)
- Gavin P Dunn
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
89
|
Wastowski IJ, Simões RT, Yaghi L, Donadi EA, Pancoto JT, Poras I, Lechapt-Zalcman E, Bernaudin M, Valable S, Carlotti CG, Flajollet S, Jensen SS, Ferrone S, Carosella ED, Kristensen BW, Moreau P. Human leukocyte antigen-G is frequently expressed in glioblastoma and may be induced in vitro by combined 5-aza-2'-deoxycytidine and interferon-γ treatments: results from a multicentric study. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:540-52. [PMID: 23219427 DOI: 10.1016/j.ajpath.2012.10.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 10/22/2012] [Accepted: 10/25/2012] [Indexed: 12/31/2022]
Abstract
Human leukocyte antigen-G (HLA-G) is a nonclassical major histocompatibility complex (MHC) class I molecule involved in immune tolerance processes, playing an important role in the maintenance of the semi-allogeneic fetus. Although HLA-G expression is restricted in normal tissues, it is broadly expressed in malignant tumors and may favor tumor immune escape. We analyzed HLA-G protein and mRNA expression in tumor samples from patients with glioblastoma collected in France, Denmark, and Brazil. We found HLA-G protein expression in 65 of 108 samples and mRNA in 20 of 21 samples. The absence of HLA-G protein expression was associated with a better long-term survival rate. The mechanisms underlying HLA-G gene expression were investigated in glioma cell lines U251MG, D247MG, and U138MG. Induction of HLA-G transcriptional activity was dependent of 5-aza-2'-deoxycytidine treatment and enhanced by interferon-γ. HLA-G protein expression was observed in U251MG cells only. These cells exhibited a permissive chromatin state at the HLA-G gene promoter and the highest levels of induced HLA-G transcriptional activity following 5-aza-2'-deoxycytidine treatment. Several antigen-presenting machinery components were up-regulated in U251MG cells after demethylating and IFN-γ treatments, suggesting an effect on the up-regulation of HLA-G cell surface expression. Therefore, because of its role in tumor tolerance, HLA-G found to be expressed in glioblastoma samples should be taken into consideration in clinical studies on the pathology and in the design of therapeutic strategies to prevent its expression in HLA-G-negative tumors.
Collapse
Affiliation(s)
- Isabela J Wastowski
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Institut des Maladies Emergentes et des Thérapies Innovantes, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Effective treatment of an orthotopic xenograft model of human glioblastoma using an EGFR-retargeted oncolytic herpes simplex virus. Mol Ther 2012; 21:561-9. [PMID: 23070115 DOI: 10.1038/mt.2012.211] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains an untreatable human brain malignancy. Despite promising preclinical studies using oncolytic herpes simplex virus (oHSV) vectors, efficacy in patients has been limited by inefficient virus replication in tumor cells. This disappointing outcome can be attributed in part to attenuating mutations engineered into these viruses to prevent replication in normal cells. Alternatively, retargeting of fully replication-competent HSV to tumor-associated receptors has the potential to achieve tumor specificity without impairment of oncolytic activity. Here, we report the establishment of an HSV retargeting system that relies on the combination of two engineered viral glycoproteins, gD and gB, to mediate highly efficient HSV infection exclusively through recognition of the abundantly expressed epidermal growth factor receptor (EGFR) on glioblastoma cells. We demonstrate efficacy in vitro and in a heterotopic tumor model in mice. Evidence for systemically administered virus homing to the tumor mass is presented. Treatment of orthotopic primary human GBM xenografts demonstrated prolonged survival with up to 73% of animals showing a complete response as confirmed by magnetic resonance imaging. Our study describes an approach to HSV retargeting that is effective in a glioma model and may be applicable to the treatment of a broad range of tumor types.
Collapse
|
91
|
Abstract
Because several antigenic peptides of human tumors that are recognized by T-lymphocytes have been identified, immune responses against cancer can now be artificially manipulated. Furthermore, since T-lymphocytes have been found to play an important role in the rejection of tumors by the host and also to have antigen-specific proliferative potentials and memory mechanisms, T-lymphocytes are thought to play a central role in cancer vaccination. Although multidisciplinary therapies have been attempted for the treatment of gliomas, the results remain unsatisfactory. For the development of new therapies against gliomas, it is required to identify tumor antigens as targets for specific immunotherapy. In this chapter, recent progress in research on glioma antigens is described.
Collapse
Affiliation(s)
- Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
92
|
Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother 2012; 62:125-35. [PMID: 22847020 PMCID: PMC3541928 DOI: 10.1007/s00262-012-1319-0] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 07/06/2012] [Indexed: 11/07/2022]
Abstract
Background This study evaluated the safety and immune responses to an autologous dendritic cell vaccine pulsed with class I peptides from tumor-associated antigens (TAA) expressed on gliomas and overexpressed in their cancer stem cell population (ICT-107). Methods TAA epitopes included HER2, TRP-2, gp100, MAGE-1, IL13Rα2, and AIM-2. HLA-A1- and/or HLA-A2-positive patients with glioblastoma (GBM) were eligible. Mononuclear cells from leukapheresis were differentiated into dendritic cells, pulsed with TAA peptides, and administered intradermally three times at two-week intervals. Results Twenty-one patients were enrolled with 17 newly diagnosed (ND-GBM) and three recurrent GBM patients and one brainstem glioma. Immune response data on 15 newly diagnosed patients showed 33 % responders. TAA expression by qRT-PCR from fresh-frozen tumor samples showed all patient tumors expressed at least three TAA, with 75 % expressing all six. Correlations of increased PFS and OS with quantitative expression of MAGE1 and AIM-2 were observed, and a trend for longer survival was observed with gp100 and HER2 antigens. Target antigens gp100, HER1, and IL13Rα2 were downregulated in recurrent tumors from 4 HLA-A2+ patients. A decrease in or absence of CD133 expression was seen in five patients who underwent a second resection. At a median follow-up of 40.1 months, six of 16 ND-GBM patients showed no evidence of tumor recurrence. Median PFS in newly diagnosed patients was 16.9 months, and median OS was 38.4 months. Conclusions Expression of four ICT-107 targeted antigens in the pre-vaccine tumors correlated with prolonged overall survival and PFS in ND-GBM patients. The goal of targeting tumor antigens highly expressed on glioblastoma cancer stem cells is supported by the observation of decreased or absent CD133 expression in the recurrent areas of gadolinium-enhanced tumors.
Collapse
|
93
|
Syed ON, Mandigo CE, Killory BD, Canoll P, Bruce JN. Cancer-testis and melanocyte-differentiation antigen expression in malignant glioma and meningioma. J Clin Neurosci 2012; 19:1016-21. [DOI: 10.1016/j.jocn.2011.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 10/18/2011] [Indexed: 11/27/2022]
|
94
|
Abstract
The outcome for patients with the most common primary brain tumor, glioblastoma multiforme (GBM), remains poor. Several immunotherapeutic approaches are actively being pursued including antibodies and cell-based therapies. While the blood-brain barrier protects brain tumor cells from therapeutic antibodies, immune cells have the ability to traverse the blood-brain barrier and migrate into GBM tumors to exert their therapeutic function. Results of Phase I clinical studies with vaccines to induce GBM-specific T cells are encouraging and Phase II clinical trials are in progress. Nonvaccine-based cell therapy for GBM has been actively explored over the last four decades. Here we will review past clinical experience with adoptive cell therapies for GBM and summarize current strategies on how to improve these approaches.
Collapse
Affiliation(s)
- K H Chow
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
95
|
Xue ZT, Sjögren HO, Salford LG, Widegren B. An epigenetic mechanism for high, synergistic expression of indoleamine 2,3-dioxygenase 1 (IDO1) by combined treatment with zebularine and IFN-γ: potential therapeutic use in autoimmune diseases. Mol Immunol 2012; 51:101-11. [PMID: 22424783 DOI: 10.1016/j.molimm.2012.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/14/2012] [Indexed: 12/21/2022]
Abstract
IDO1 can be induced by interferon gamma (IFN-γ) in multiple cell types. We have earlier described that the DNA methyltransferase inhibitor zebularine also induces IDO1 in several rat cell clones. We now describe a synergistic induction of IDO1 expression by IFN-γ and zebularine. To elucidate the mechanism of the IDO1 induction we have studied the methylation status in the promoter region of the IDO1 gene from both human monocytic THP-1 cells and H1D2 rat colon cancer cells. Interestingly, the IDO1 promoter is hypermethylated and IFN-γ is shown to induce a significant demethylation. The synergism in effect of zebularine and IFN-γ on IDO1 expression is paralleled by a similar synergistic effect on expression of two other IFN-γ-responsive genes, the transcription factors STAT1 and IRF1 with binding sites in the IDO1 promoter region. The demonstrated synergistic activation of IDO1 expression has implications in relation to therapeutic induction of immunosuppression in autoimmunity and chronic inflammation.
Collapse
Affiliation(s)
- Zhong-Tian Xue
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
96
|
Mahzouni P, Movahedipour M. An immunohistochemical study of HER2 expression in meningioma and its correlation with tumor grade. Pathol Res Pract 2012; 208:221-4. [PMID: 22420924 DOI: 10.1016/j.prp.2012.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 12/17/2011] [Accepted: 01/30/2012] [Indexed: 10/28/2022]
Abstract
Meningiomas account for about 15-30% of all primary intracranial tumors. According to the 2007 WHO classification, meningiomas are divided into three grades (I, II and III). Recurrence is an issue following surgical treatment of meningioma, especially in grades II and III. HER2 (also known as erbB-2) is a 185-kD transmembrane glycoprotein with tyrosine kinase activity. HER2 is expressed in some human malignancies and can be a potential target for therapeutic intervention with selective inhibitors. There are only a few studies on the relationship between meningioma and HER2 expression, and the results are different as well. The aim of this study was to determine this relationship. Seventy-two paraffin blocks of meningioma were selected randomly, and immunohistochemical staining was then performed for each specimen. Thirty-one of the 72 meningiomas were HER2-positive. HER2 expression was observed in 11 (55%) of the 20 grade II/III, and 20 (38.5%) of the 52 grade I meningiomas. Consequently, HER2 expression was detected in 43% of meningiomas. No significant difference was seen between grade I and grade II/III meningiomas, primary and recurrent tumors, and males and females from the point of view of HER2 expression.
Collapse
Affiliation(s)
- Parvin Mahzouni
- Department of Pathology, Medical University of Isfahan, Isfahan, Iran
| | | |
Collapse
|
97
|
Cellular-based immunotherapies for patients with glioblastoma multiforme. Clin Dev Immunol 2012; 2012:764213. [PMID: 22474481 PMCID: PMC3299309 DOI: 10.1155/2012/764213] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 12/08/2011] [Indexed: 12/30/2022]
Abstract
Treatment of patients with glioblastoma multiforme (GBM) remains to be a challenge with a median survival of 14.6 months following diagnosis. Standard treatment options include surgery, radiation therapy, and systemic chemotherapy with temozolomide. Despite the fact that the brain constitutes an immunoprivileged site, recent observations after immunotherapies with lysate from autologous tumor cells pulsed on dendritic cells (DCs), peptides, protein, messenger RNA, and cytokines suggest an immunological and even clinical response from immunotherapies. Given this plethora of immunomodulatory therapies, this paper gives a structure overview of the state-of-the art in the field. Particular emphasis was also put on immunogenic antigens as potential targets for a more specific stimulation of the immune system against GBM.
Collapse
|
98
|
Ikeda H, Shiku H. Antigen-receptor gene-modified T cells for treatment of glioma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:202-15. [PMID: 22639170 DOI: 10.1007/978-1-4614-3146-6_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Immunological effector cells and molecules have been shown to access intracranial tumor sites despite the existence of blood brain barrier (BBB) or immunosuppressive mechanisms associated with brain tumors. Recent progress in T-cell biology and tumor immunology made possible to develop strategies of tumor-associated antigen-specific immunotherapeutic approaches such as vaccination with defined antigens and adoptive T-cell therapy with antigen-specific T cells including gene-modified T cells for the treatment of patients with brain tumors. An array of recent reports on the trials of active and passive immunotherapy for patients with brain tumors have documented safety and some preliminary clinical efficacy, although the ultimate judgment for clinical benefits awaits rigorous evaluation in trials of later phases. Nevertheless, treatment with lymphocytes that are engineered to express tumor-specific receptor genes is a promising immunotherapy against glioma, based on the significant efficacy reported in the trials for patients with other types of malignancy. Overcoming the relative difficulty to apply immunotherapeutic approach to intracranial region, current advances in the understanding of human tumor immunology and the gene-therapy methodology will address the development of effective immunotherapy of brain tumors.
Collapse
Affiliation(s)
- Hiroaki Ikeda
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan.
| | | |
Collapse
|
99
|
Longo SL, Padalino DJ, McGillis S, Petersen K, Schirok H, Politz O, Canute GW, Post DE. Bay846, a new irreversible small molecule inhibitor of EGFR and Her2, is highly effective against malignant brain tumor models. Invest New Drugs 2011; 30:2161-72. [PMID: 22203214 DOI: 10.1007/s10637-011-9784-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 12/09/2011] [Indexed: 12/22/2022]
Abstract
The epidermal growth factor receptor (EGFR) pathway is aberrantly activated in tumors and plays a key role in promoting tumor growth. Small molecule inhibitors which bind reversibly to EGFR have demonstrated limited clinical activity. Thus, there is a continued need to develop novel EGFR inhibitors with improved anti-tumor activity. Bay846 is a newly developed small molecule inhibitor that binds irreversibly to the tyrosine kinase domains of EGFR and Her2. The in vitro and in vivo efficacy of Bay846 was tested using a panel of nine human malignant brain tumor (glioma) models. Lapatinib, a reversible inhibitor of EGFR and Her2, was included for comparison. Six glioma cell lines were sensitive to Bay846 treatment. Bay846 strongly suppressed tumor cell growth in vitro by inducing cell lysis/death rather than cell cycle arrest. Consistent with this, Bay846 had potent anti-tumor activity which led to regressions in tumor size. The active, phosphorylated form of EGFR was reduced by Bay846 treatment in vitro and in tumors. Importantly, the efficacy of Bay846 was significantly greater than lapatinib in all assays. Bay846-sensitivity was associated with expression of a wild-type PTEN in conjunction with high levels of an oncogenic EGFR variant (A289V or EGFRvIII). These studies demonstrate that targeting the EGFR pathway with the irreversible inhibitor Bay846 has great potential to increase the efficacy of this cancer therapy.
Collapse
Affiliation(s)
- Sharon L Longo
- Department of Neurosurgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Reszeć J, Bernaczyk PS, Milewski R, Chyczewski L, Mariak Z. c-erbB-2 protein expression in astrocytic tumors of the brain. Med Sci Monit 2011; 17:BR216-220. [PMID: 21804458 PMCID: PMC3539622 DOI: 10.12659/msm.881900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background Astrocytic tumors are the primary brain tumors, which often progress to glioblastoma, a highly malignant neoplasm of the central nervous system. There is much new data regarding to the formation and progression of these tumors; however, glioblastoma remains one of the most fatal neoplasms in humans. The aim of the study was to evaluate the role of c-erbB-2 protein expression in various groups of astrocytic tumors. Material/Methods 65 cases of astrocytic tumors were divided into 3 groups: diffuse astrocytoma (group I; n=17 cases), anaplastic astrocytoma (group II; n=23 cases) and glioblastoma (group III; n=25 cases). C-erbB-2 protein expression was estimated semiquantitatively on immunohistochemically stained tissue sections using antibodies against c-erbB-2 protein. Statistical analysis was performed in all examined groups. Results The c-erbB-2 protein expression was observed in 15 out of 17 cases (88.3%) in group I, 22 out of 25 cases (88%) cases in group II, and in 19 out of 23 cases (82.6%) in group III. There were no statistically significant differences between the examined groups. The strongest c-erbB-2 immunoexpression was observed in low grade astrocytomas (diffuse astrocytomas G2); in the glioblastoma group the c-erbB-2 protein expression was weak and 17.4% of cases were negative. Conclusions C-erbB-2 protooncogene alteration is an early phenomenon in glial tumor development and progression.
Collapse
Affiliation(s)
- Joanna Reszeć
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland.
| | | | | | | | | |
Collapse
|