1
|
Van Gool SW, Van de Vliet P, Kampers LFC, Kosmal J, Sprenger T, Reich E, Schirrmacher V, Stuecker W. Methods behind oncolytic virus-based DC vaccines in cancer: Toward a multiphase combined treatment strategy for Glioblastoma (GBM) patients. Methods Cell Biol 2023; 183:51-113. [PMID: 38548421 DOI: 10.1016/bs.mcb.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Glioblastoma (GBM) remains an orphan cancer disease with poor outcome. Novel treatment strategies are needed. Immunotherapy has several modes of action. The addition of active specific immunotherapy with dendritic cell vaccines resulted in improved overall survival of patients. Integration of DC vaccination within the first-line combined treatment became a challenge, and immunogenic cell death immunotherapy during chemotherapy was introduced. We used a retrospective analysis using real world data to evaluate the complex combined treatment, which included individualized multimodal immunotherapy during and after standard of care, and which required adaptations during treatment, and found a further improvement of overall survival. We also discuss the use of real world data as evidence. Novel strategies to move the field of individualized multimodal immunotherapy forward for GBM patients are reviewed.
Collapse
Affiliation(s)
| | | | | | | | | | - Ella Reich
- Immun-onkologisches Zentrum Köln, Cologne, Germany
| | | | | |
Collapse
|
2
|
Dain L, Zhu G. Nucleic acid immunotherapeutics and vaccines: A promising approach to glioblastoma multiforme treatment. Int J Pharm 2023; 638:122924. [PMID: 37037396 PMCID: PMC10194422 DOI: 10.1016/j.ijpharm.2023.122924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023]
Abstract
Glioblastoma multiforme (GBM) is a deadly and difficult to treat primary brain tumor for which satisfactory therapeutics have yet to be discovered. While cancer immunotherapeutics, such as immune checkpoint inhibitors, have successfully improved the treatment of some other types of cancer, the poorly immunogenic GBM tumor cells and the immunosuppressive GBM tumor microenvironment have made it difficult to develop GBM immunotherapeutics. Nucleic acids therapeutics and vaccines, particularly those of mRNA, have become a popular field of research in recent years. This review presents the progress of nucleic acid therapeutics and vaccines for GBM and briefly covers some representative delivery methods of nucleic acids to the central nervous system (CNS) for GBM therapy.
Collapse
Affiliation(s)
- Lauren Dain
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy; The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Guizhi Zhu
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy; The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
3
|
Bausart M, Rodella G, Dumont M, Ucakar B, Vanvarenberg K, Malfanti A, Préat V. Combination of local immunogenic cell death-inducing chemotherapy and DNA vaccine increases the survival of glioblastoma-bearing mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102681. [PMID: 37105343 DOI: 10.1016/j.nano.2023.102681] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy efficacy as monotherapy is negligible for glioblastoma (GBM). We hypothesized that combining therapeutic vaccination using a plasmid encoding an epitope derived from GBM-associated antigen (pTOP) with local delivery of immunogenic chemotherapy using mitoxantrone-loaded PEGylated PLGA-based nanoparticles (NP-MTX) would improve the survival of GBM-bearing mice by stimulating an antitumor immune response. We first proved that MTX retained its ability to induce cytotoxicity and immunogenic cell death of GBM cells after encapsulation. Intratumoral delivery of MTX or NP-MTX increased the frequency of IFN-γ-secreting CD8 T cells. NP-MTX mixed with free MTX in combination with pTOP DNA vaccine increased the median survival of GL261-bearing mice and increased M1-like macrophages in the brain. The addition of CpG to this combination abolished the survival benefit but led to increased M1 to M2 macrophage ratio and IFN-γ-secreting CD4 T cell frequency. These results highlight the benefits of combination strategies to potentiate immunotherapy and improve GBM outcome.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Mathilde Dumont
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Bernard Ucakar
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| |
Collapse
|
4
|
Combination of DNA Vaccine and Immune Checkpoint Blockades Improves the Immune Response in an Orthotopic Unresectable Glioblastoma Model. Pharmaceutics 2022; 14:pharmaceutics14051025. [PMID: 35631612 PMCID: PMC9145362 DOI: 10.3390/pharmaceutics14051025] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
Combination immunotherapy has emerged as a promising strategy to increase the immune response in glioblastoma (GBM) and overcome the complex immunosuppression occurring in its microenvironment. In this study, we hypothesized that combining DNA vaccines—to stimulate a specific immune response—and dual immune checkpoint blockade (ICB)—to decrease the immunosuppression exerted on T cells—will improve the immune response and the survival in an orthotopic unresectable GL261 model. We first highlighted the influence of the insertion position of a GBM epitope sequence in a plasmid DNA vaccine encoding a vesicular stomatitis virus glycoprotein (VSV-G) (here referred to as pTOP) in the generation of a specific and significant IFN-γ response against the GBM antigen TRP2 by inserting a CD8 epitope sequence in specific permissive sites. Then, we combined the pTOP vaccine with anti-PD-1 and anti-CTLA-4 ICBs. Immune cell analysis revealed an increase in effector T cell to Treg ratios in the spleens and an increase in infiltrated IFN-γ-secreting CD8 T cell frequency in the brains following combination therapy. Even if the survival was not significantly different between dual ICB and combination therapy, we offer a new immunotherapeutic perspective by improving the immune landscape in an orthotopic unresectable GBM model.
Collapse
|
5
|
Saha S, Venu Y, Bhattacharya D, Kompella SD, Madhusudana K, Chakravarty S, Ramakrishna S, Chaudhuri A. Combating Established Mouse Glioblastoma through Nicotinylated-Liposomes-Mediated Targeted Chemotherapy in Combination with Dendritic-Cell-Based Genetic Immunization. ACTA ACUST UNITED AC 2017; 1:e1600009. [DOI: 10.1002/adbi.201600009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/30/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Soumen Saha
- Biomaterials Group; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
- Academy of Scientific & Innovative Research (AcSIR); 2 Rafi Marg New Delhi 110001 India
| | - Yakati Venu
- Biomaterials Group; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
- Academy of Scientific & Innovative Research (AcSIR); 2 Rafi Marg New Delhi 110001 India
| | - Dwaipayan Bhattacharya
- Chemical Biology Division; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
| | - Shobha Deepthi Kompella
- Biomaterials Group; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
| | - Kuncha Madhusudana
- Medicinal Chemistry and Pharmacology Division; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
| | - Sumana Chakravarty
- Chemical Biology Division; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
| | - Sistla Ramakrishna
- Medicinal Chemistry and Pharmacology Division; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
| | - Arabinda Chaudhuri
- Biomaterials Group; CSIR-Indian Institute of Chemical Technology; Uppal Road, Tarnaka Hyderabad 500007 Telangana India
- Academy of Scientific & Innovative Research (AcSIR); 2 Rafi Marg New Delhi 110001 India
| |
Collapse
|
6
|
Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A, Chlichlia K. DNA vaccines to attack cancer: Strategies for improving immunogenicity and efficacy. Pharmacol Ther 2016; 165:32-49. [DOI: 10.1016/j.pharmthera.2016.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
7
|
Kalyanasundram J, Chia SL, Song AAL, Raha AR, Young HA, Yusoff K. Surface display of glycosylated Tyrosinase related protein-2 (TRP-2) tumour antigen on Lactococcus lactis. BMC Biotechnol 2015; 15:113. [PMID: 26715153 PMCID: PMC4696278 DOI: 10.1186/s12896-015-0231-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/22/2015] [Indexed: 01/27/2023] Open
Abstract
Background The exploitation of the surface display system of food and commensal lactic acid bacteria (LAB) for bacterial, viral, or protozoan antigen delivery has received strong interest recently. The Generally Regarded as Safe (GRAS) status of the Lactococcus lactis coupled with a non-recombinant strategy of in-trans surface display, provide a safe platform for therapeutic drug and vaccine development. However, production of therapeutic proteins fused with cell-wall anchoring motifs is predominantly limited to prokaryotic expression systems. This presents a major disadvantage in the surface display system particularly when glycosylation has been recently identified to significantly enhance epitope presentation. In this study, the glycosylated murine Tyrosinase related protein-2 (TRP-2) with the ability to anchor onto the L. lactis cell wall was produced in suspension adapted Chinese Hamster Ovary (CHO-S) cells by expressing TRP-2 fused with cell wall anchoring LysM motif (cA) at the C-terminus. Results A total amount of 33 μg of partially purified TRP-2-cA from ~6.0 g in wet weight of CHO-S cells was purified by His-tag affinity chromatography. The purified TRP-2-cA protein was shown to be N-glycosylated and successfully anchored to the L. lactis cell wall. Conclusions Thus cell surface presentation of glycosylated mammalian antigens may now permit development of novel and inexpensive vaccine platforms.
Collapse
Affiliation(s)
- Jeevanathan Kalyanasundram
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia.
| | - Suet Lin Chia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia.
| | - Adelene Ai-Lian Song
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia.
| | - Abdul Rahim Raha
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia. .,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia.
| | - Howard A Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor Darul Ehsan, Malaysia. .,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
8
|
Fenton KE, Martirosyan NL, Abdelwahab MG, Coons SW, Preul MC, Scheck AC. In vivo visualization of GL261-luc2 mouse glioma cells by use of Alexa Fluor-labeled TRP-2 antibodies. Neurosurg Focus 2014; 36:E12. [PMID: 24484250 DOI: 10.3171/2013.12.focus13488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT For patients with glioblastoma multiforme, median survival time is approximately 14 months. Longer progression-free and overall survival times correlate with gross-total resection of tumor. The ability to identify tumor cells intraoperatively could result in an increased percentage of tumor resected and thus increased patient survival times. Available labeling methods rely on metabolic activity of tumor cells; thus, they are more robust in high-grade tumors, and their utility in low-grade tumors and metastatic tumors is not clear. The authors demonstrate intraoperative identification of tumor cells by using labeled tumor-specific antibodies. METHODS GL261 mouse glioma cells exhibit high expression of a membrane-bound protein called second tyrosinase-related protein (TRP-2). The authors used these cells to establish an intracranial, immunocompetent model of malignant glioma. Antibodies to TRP-2 were labeled by using Alexa Fluor 488 fluorescent dye and injected into the tail vein of albino C57BL/6 mice. After 24 hours, a craniotomy was performed and the tissue was examined in vivo by using an Optiscan 5.1 handheld portable confocal fiber-optic microscope. Tissue was examined ex vivo by using a Pascal 5 scanning confocal microscope. RESULTS Labeled tumor cells were visible in vivo and ex vivo under the respective microscopes. CONCLUSIONS Fluorescently labeled tumor-specific antibodies are capable of binding and identifying tumor cells in vivo, accurately and specifically. The development of labeled markers for the identification of brain tumors will facilitate the use of intraoperative fluorescence microscopy as a tool for increasing the extent of resection of a broad variety of intracranial tumors.
Collapse
|
9
|
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: 1.9] [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
|
10
|
Kang TH, Mao CP, La V, Chen A, Hung CF, Wu TC. Innovative DNA vaccine to break immune tolerance against tumor self-antigen. Hum Gene Ther 2013; 24:181-8. [PMID: 23268564 DOI: 10.1089/hum.2012.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vaccination is, in theory, a safe and effective approach for controlling disseminated or metastatic cancer due to the specificity of the mammalian immune system, yet its success in the clinic has been hampered thus far by the problem of immune tolerance to tumor self-antigen. Here we describe a DNA vaccination strategy that is able to control cancer by overcoming immune tolerance to tumor self-antigen. We engineered a DNA construct encoding a dimeric form of a secreted single-chain trimer of major histocompatibility complex class I heavy chain, β2-microglobulin, and peptide antigen linked to immunoglobulin G (SCT-Ag/IgG). The chimeric protein was able to bind to antigen-specific CD8(+) T cells with nearly 100% efficiency and strongly induce their activation and proliferation. In addition, the chimeric protein was able to coat professional antigen-presenting cells through the F(c) receptor to activate antigen-specific CD8(+) T cells. Furthermore, intradermal vaccination with DNA-encoding SCT-Ag/IgG could generate significant numbers of cytotoxic effector T cells against tumor self-antigen and leads to successful therapeutic outcomes in a preclinical model of metastatic melanoma. Our data suggest that the DNA vaccine strategy described in the current study is able to break immune tolerance against endogenous antigen from melanoma and result in potent therapeutic antitumor effects. Such strategy may be used in other antigenic systems for the control of infections and/or cancers.
Collapse
Affiliation(s)
- Tae Heung Kang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | | | | | | | | | | |
Collapse
|
11
|
Ginzkey C, Eicker S, Marget M, Krause J, Brecht S, Westphal M, Hugo HH, Mehdorn M, Steinmann J, Hamel W. Incomplete tumour control following DNA vaccination against rat gliomas expressing a model antigen. Acta Neurochir (Wien) 2013; 155:51-8; discussion 59. [PMID: 23132370 PMCID: PMC3535398 DOI: 10.1007/s00701-012-1526-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 10/11/2012] [Indexed: 11/26/2022]
Abstract
Background Vaccination against tumour-associated antigens is one approach to elicit anti-tumour responses. We investigated the effect of polynucleotide (DNA) vaccination using a model antigen (E. coli lacZ) in a syngeneic gliosarcoma model (9L). Methods Fisher 344 rats were vaccinated thrice by intramuscular injection of a lacZ-encoding or a control plasmid in weekly intervals. One week after the last vaccination, lacZ-expressing 9L cells were implanted into the striatum. Results After 3 weeks, in lacZ-vaccinated animals the tumours were significantly smaller than in control-vaccinated animals. In cytotoxic T cell assays lysis rates of >50 % could only be observed in a few of the lacZ-vaccinated animals. This response was directed against lacZ-expressing and parental 9L cells but not against syngeneic MADB 106 adenocarcinoma cells. In Elispot assays interferon-γ production was observed upon stimulation with 9LlacZ and 9L wild-type but not MADB 106 cells. This response was higher for lacZ-immunized animals. All animals revealed dense infiltrates with CD8+ lymphocytes and, to a lesser extent, with NK cells. CD25-staining indicated cells possibly associated with the maintenance of peripheral tolerance to self-antigens. All tumours were densely infiltrated by microglia consisting mostly of ramified cells. Only focal accumulation of macrophage-like cells expressing ED1, a marker for phagocytic activity, was observed. Conclusion Prophylactic DNA vaccination resulted in effective but incomplete suppression of brain tumour formation. Mechanisms other than cytotoxic T cell responses as measured in the generally used in vitro assays appear to play a role in tumour suppression.
Collapse
Affiliation(s)
- Christian Ginzkey
- Department of Neurosurgery, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Department of Oto-Rhino-Laryngology, Julius-Maximilian-University, Würzburg, Germany
| | - Sven Eicker
- Department of Neurosurgery, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Department of Neurosurgery, Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias Marget
- Institute of Immunology, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jörg Krause
- Department of Neurosurgery, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Stefan Brecht
- Institute of Pharmacology, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Klinik für Neurochirurgie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Heinz-Hermann Hugo
- Department of Neurosurgery, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Maximilian Mehdorn
- Department of Neurosurgery, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jörg Steinmann
- Institute of Immunology, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Wolfgang Hamel
- Department of Neurosurgery, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Klinik für Neurochirurgie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
12
|
Takahashi S, Fusaki N, Ohta S, Iwahori Y, Iizuka Y, Inagawa K, Kawakami Y, Yoshida K, Toda M. Downregulation of KIF23 suppresses glioma proliferation. J Neurooncol 2011; 106:519-29. [PMID: 21904957 DOI: 10.1007/s11060-011-0706-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 08/18/2011] [Indexed: 12/18/2022]
Abstract
To identify therapeutic molecular targets for glioma, we performed modified serological identification of antigens by recombinant complementary DNA (cDNA) expression cloning using sera from a mouse glioma model. Two clones, kinesin family member 23 (Kif23) and structural maintenance of chromosomes 4 (Smc4), were identified as antigens through immunological reaction with sera from mice harboring synergic GL261 mouse glioma and intratumoral inoculation with a mutant herpes simplex virus. The human Kif23 homolog KIF23 is a nuclear protein that localizes to the interzone of mitotic spindles, acting as a plus-end-directed motor enzyme that moves antiparallel microtubules in vitro. Expression analysis revealed a higher level of KIF23 expression in glioma tissues than in normal brain tissue. The introduction of small interfering RNA (siRNA) targeting KIF23 into two different glioma cell lines, U87MG and SF126, downregulated KIF23 expression, which significantly suppressed glioma cell proliferation in vitro. KIF23 siRNA-treated glioma cells exhibited larger cell bodies with two or more nuclei compared with control cells. In vivo analysis using mouse xenograft showed that KIF23 siRNA/DNA chimera-treated tumors were significantly smaller than tumors treated with control siRNA/DNA chimera. Taken together, our results indicate that downregulation of KIF23 decreases proliferation of glioma cells and that KIF23 may be a novel therapeutic target in malignant glioma.
Collapse
Affiliation(s)
- Satoshi Takahashi
- Department of Neurosurgery, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
CD4+ and CD8+ T cells can act separately in tumour rejection after immunization with murine pneumotropic virus chimeric Her2/neu virus-like particles. PLoS One 2010; 5:e11580. [PMID: 20657846 PMCID: PMC2906518 DOI: 10.1371/journal.pone.0011580] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 06/10/2010] [Indexed: 01/21/2023] Open
Abstract
Background Immunization with murine pneumotropic virus virus-like particles carrying Her2/neu (Her2MPtVLPs) prevents tumour outgrowth in mice when given prophylactically, and therapeutically if combined with the adjuvant CpG. We investigated which components of the immune system are involved in tumour rejection, and whether long-term immunological memory can be obtained. Methodology and Results During the effector phase in BALB/c mice, only depletion of CD4+ and CD8+ in combination, with or without NK cells, completely abrogated tumour protection. Depletion of single CD4+, CD8+ or NK cell populations only had minor effects. During the immunization/induction phase, combined depletion of CD4+ and CD8+ cells abolished protection, while depletion of each individual subset had no or negligible effect. When tumour rejection was studied in knock-out mice with a C57Bl/6 background, protection was lost in CD4−/−CD8−/− and CD4−/−, but not in CD8−/− mice. In contrast, when normal C57Bl/6 mice were depleted of different cell types, protection was lost irrespective of whether only CD4+, only CD8+, or CD4+ and CD8+ cells in combination were eradicated. No anti-Her2/neu antibodies were detected but a Her2/neu-specific IFNγ response was seen. Studies of long-term memory showed that BALB/c mice could be protected against tumour development when immunized together with CpG as long as ten weeks before challenge. Conclusion Her2MPtVLP immunization is efficient in stimulating several compartments of the immune system, and induces an efficient immune response including long-term memory. In addition, when depleting mice of isolated cellular compartments, tumour protection is not as efficiently abolished as when depleting several immune compartments together.
Collapse
|
14
|
Bridle BW, Li J, Jiang S, Chang R, Lichty BD, Bramson JL, Wan Y. Immunotherapy can reject intracranial tumor cells without damaging the brain despite sharing the target antigen. THE JOURNAL OF IMMUNOLOGY 2010; 184:4269-75. [PMID: 20237288 DOI: 10.4049/jimmunol.0901447] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although vaccines targeting tissue differentiation Ags represent a promising strategy for cancer immunotherapy, the risk of triggering autoimmune damage to normal tissues remains to be determined. Immunizing against a melanoma-associated Ag, dopachrome tautomerase (DCT), which normal melanocytes and glial cells also express, allowed concurrent analysis of autoimmune consequences in multiple tissues. We show that vaccination with recombinant adenovirus expressing DCT elicited a strong CTL response in C57BL/6 mice, leading to protection against intracranial challenge with B16-F10 melanoma cells. Both histological analysis and behavioral testing indicated that there was no evidence of neuropathology in vaccinated animals and long-term survivors. Although vitiligo or demyelination could be induced by additional stimuli (i.e., surgery or inflammation) in DCT-vaccinated mice, it did not extend beyond the inflammatory area, suggesting that there is self-regulatory negative feedback in normal tissues. These results demonstrate that it is possible to vaccinate against a tumor embedded in a vital organ that shares the target Ag.
Collapse
Affiliation(s)
- Byram W Bridle
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
15
|
Ghulam Muhammad AKM, Candolfi M, King GD, Yagiz K, Foulad D, Mineharu Y, Kroeger KM, Treuer KA, Nichols WS, Sanderson NS, Yang J, Khayznikov M, Van Rooijen N, Lowenstein PR, Castro MG. Antiglioma immunological memory in response to conditional cytotoxic/immune-stimulatory gene therapy: humoral and cellular immunity lead to tumor regression. Clin Cancer Res 2009; 15:6113-27. [PMID: 19789315 DOI: 10.1158/1078-0432.ccr-09-1087] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma multiforme is a deadly primary brain cancer. Because the tumor kills due to recurrences, we tested the hypothesis that a new treatment would lead to immunological memory in a rat model of recurrent glioblastoma multiforme. EXPERIMENTAL DESIGN We developed a combined treatment using an adenovirus (Ad) expressing fms-like tyrosine kinase-3 ligand (Flt3L), which induces the infiltration of immune cells into the tumor microenvironment, and an Ad expressing herpes simplex virus-1-thymidine kinase (TK), which kills proliferating tumor cells in the presence of ganciclovir. RESULTS This treatment induced immunological memory that led to rejection of a second glioblastoma multiforme implanted in the contralateral hemisphere and of an extracranial glioblastoma multiforme implanted intradermally. Rechallenged long-term survivors exhibited anti-glioblastoma multiforme-specific T cells and displayed specific delayed-type hypersensitivity. Using depleting antibodies, we showed that rejection of the second tumor was dependent on CD8(+) T cells. Circulating anti-glioma antibodies were observed when glioblastoma multiforme cells were implanted intradermally in naïve rats or in long-term survivors. However, rats bearing intracranial glioblastoma multiforme only exhibited circulating antitumoral antibodies upon treatment with Ad-Flt3L + Ad-TK. This combined treatment induced tumor regression and release of the chromatin-binding protein high mobility group box 1 in two further intracranial glioblastoma multiforme models, that is, Fisher rats bearing intracranial 9L and F98 glioblastoma multiforme cells. CONCLUSIONS Treatment with Ad-Flt3L + Ad-TK triggered systemic anti-glioblastoma multiforme cellular and humoral immune responses, and anti-glioblastoma multiforme immunological memory. Release of the chromatin-binding protein high mobility group box 1 could be used as a noninvasive biomarker of therapeutic efficacy for glioblastoma multiforme. The robust treatment efficacy lends further support to its implementation in a phase I clinical trial.
Collapse
Affiliation(s)
- A K M Ghulam Muhammad
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Maes W, Deroose C, Reumers V, Krylyshkina O, Gijsbers R, Baekelandt V, Ceuppens J, Debyser Z, Van Gool SW. In vivo bioluminescence imaging in an experimental mouse model for dendritic cell based immunotherapy against malignant glioma. J Neurooncol 2008; 91:127-39. [PMID: 18787761 DOI: 10.1007/s11060-008-9691-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 08/25/2008] [Indexed: 02/07/2023]
Abstract
The value of bioluminescence imaging (BLI) for experimental cancer models has become firmly established. We applied BLI to the GL261 glioma model in the context of dendritic cell (DC) immunotherapy. Initial validation revealed robust linear correlations between in vivo, ex vivo and in vitro luciferase activity measurements. Ex vivo BLI demonstrated midline crossing and leakage of tumor cells. Orthotopically challenged mice followed with BLI showed an initial adaptation phase, after which imaging data correlated linearly with stereologically determined tumor dimensions. Transition from healthy to moribund state corresponded with an increasing in vivo flux but the onset of neurological deficit was clearly delayed compared to the onset of in vivo flux increase. BLI was implemented in prophylactic immunotherapy and imaging data were prognostic for therapy outcome. Three distinct response patterns were detected. Our data underscore the feasibility of in vivo BLI in an experimental immunotherapeutic setting in the GL261 glioma model.
Collapse
Affiliation(s)
- W Maes
- Department of Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Grauer OM, Sutmuller RPM, van Maren W, Jacobs JFM, Bennink E, Toonen LWJ, Nierkens S, Adema GJ. Elimination of regulatory T cells is essential for an effective vaccination with tumor lysate-pulsed dendritic cells in a murine glioma model. Int J Cancer 2008; 122:1794-802. [PMID: 18076066 DOI: 10.1002/ijc.23284] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Both melanoma and glioma cells are of neuroectodermal origin and share common tumor associated antigens. In this article, we report that the melanocyte differentiation antigen TRP2 (tyrosinase-related protein 2) is not predominantly involved in the tumor rejection of a syngeneic murine glioma. Although GL261 glioma cells endogenously expressed TRP2 and were lysed by TRP2 specific cytotoxic T cells (CTLs) in vitro, vaccinations with TRP2 peptide-pulsed dendritic cells (DCs) could only induce minor antiglioma responses in a prophylactic setting and failed to work in a stringent setting where vaccine and tumor were administered on the same day. Further analysis revealed that TRP2 is not recognized by bulk CTLs after depletion of regulatory T cells which results in tumor rejections in vivo. In contrast to TRP2 peptide-pulsed DC, tumor lysate-pulsed DCs were more potent as a vaccine and completely protected mice from tumor outgrowth in a prophylactic setting. However, the vaccine efficacy of tumor lysate-pulsed DC was not sufficient to prevent the tumor outgrowth when tumors were inoculated the same day. In this case, Treg depletion before vaccination was essential to boost antiglioma immune responses leading to the rejection of 80% of the mice and long-term immunity. Therefore, we conclude that counteracting the immunosuppressive glioma tumor environment via depletion of regulatory T cells is a prerequisite for successful eradication of gliomas after targeting multiple tumor antigens by using tumor lysate-pulsed DCs as a vaccine in a more stringent setting.
Collapse
Affiliation(s)
- Oliver M Grauer
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Iizuka Y, Kojima H, Kobata T, Kawase T, Kawakami Y, Toda M. Identification of a glioma antigen, GARC-1, using cytotoxic T lymphocytes induced by HSV cancer vaccine. Int J Cancer 2006; 118:942-9. [PMID: 16152596 DOI: 10.1002/ijc.21432] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Despite several ongoing clinical trials of immunotherapies against glioma, few glioma-specific antigens recognized by cytotoxic T lymphocytes (CTLs) have been identified. We recently demonstrated that intratumoral inoculation with herpes simplex virus (HSV) as a cancer vaccine activates tumor-specific CTLs. To identify glioma antigens recognized by CTLs, we used the HSV cancer vaccine to vaccinate mice harboring a syngeneic mouse glioma cell line, GL261. From the splenocytes of the immunized mice, we generated an H-2Db-restricted CTL line, GCL-1, that was specific for GL261. Then, a cDNA expression library generated from GL261 was screened with GCL-1, and a new gene encoding glioma antigen, GARC-1, was isolated. Sequence analysis revealed that the GARC-1 gene isolated from GL261 had a point mutation causing an amino acid change (Asp to Asn at position 81). T-cell epitope analysis revealed that the mutated peptide GARC-1(77-85) (AALLNKLYA) but not the wild-type peptide (AALLDKLYA), was recognized by GCL-1. These results suggest that HSV cancer vaccination may be a useful method for inducing tumor-specific CTLs and identifying tumor antigens. Furthermore, this GL261/GARC-1 murine glioma model may be useful for the development of immunotherapy for brain tumors.
Collapse
Affiliation(s)
- Yukihiko Iizuka
- Neuroimmunology Research Group, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Affiliation(s)
- Min Yu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| | - Olivera J. Finn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| |
Collapse
|
20
|
Prins RM, Bruhn KW, Craft N, Lin JW, Kim CH, Odesa SK, Miller JF, Liau LM. Central Nervous System Tumor Immunity Generated by a Recombinant Listeria monocytogenes Vaccine Targeting Tyrosinase Related Protein-2 and Real-Time Imaging of Intracranial Tumor Burden. Neurosurgery 2006; 58:169-78; discussion 169-78. [PMID: 16385341 DOI: 10.1227/01.neu.0000192367.29047.64] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Previously, we demonstrated that a recombinant Listeria monocytogenes (rLM) vector encoding the melanoma-associated antigen, tyrosinase related protein (TRP)-2, could successfully treat subcutaneous B16 melanomas. The purpose of the present study was twofold: 1) to test whether this rLM-nucleoprotein (NP)/TRP-2 could generate antitumor immunity to a B16 tumor challenge in the immunologically privileged central nervous system (CNS) and 2) to develop a noninvasive imaging modality to monitor tumor progression in the brain after immunotherapy. METHODS Mice were vaccinated with either a control rLM strain expressing only a viral antigen (rLM-NP) or a strain expressing both the viral epitope and TRP-2 (rLM-NP/TRP-2). These mice were then analyzed for their ability to mount tumor-specific T-cell responses, to generate protective antitumor immunity to a CNS tumor challenge, and for the localization of T cells at the tumor site. To noninvasively measure tumor growth within the CNS in vivo, we developed a B16 cell line expressing firefly luciferase that could be readily detected via bioluminescent imaging. RESULTS Vaccination with rLM-NP/TRP-2 induced a robust, tumor-specific CD8 T-cell response to the dominant cytotoxic T lymphocyte epitope of TRP-2 and selective interferon-gamma secretion when cocultured with B16 melanoma cells in vitro. Significant decreases in CNS tumor sizes were easily visualized in mice vaccinated with rLM-NP/TRP-2 compared with mice that received a control rLM expressing the NP epitope alone (rLM-NP). The subsequent decreased tumor size and extension of survival induced by rLM-NP/TRP-2 was similarly associated with an early increase of tumor infiltrating T cells. CONCLUSION The ability to treat tumors arising within the CNS is difficult because of the nature of the anatomic confines of the brain and a microenvironment that may not promote immune responsiveness. These studies describe an in vivo bioluminescent imaging system to monitor CNS tumor growth in mice, which we successfully used to document decreased intracranial tumor progression and size after vaccination with rLM-NP/TRP-2. The results suggest that metastatic tumors in the CNS can be targeted immunotherapeutically without overt autoimmune toxicity.
Collapse
Affiliation(s)
- Robert M Prins
- Division of Neurosurgery, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Kim JH, Chen J, Majumder N, Lin H, Falo LD, You Z. ‘Survival gene’ Bcl-xl potentiates DNA-raised antitumor immunity. Gene Ther 2005; 12:1517-25. [PMID: 16052205 DOI: 10.1038/sj.gt.3302584] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
T-cell priming is strongly affected by the longevity of antigen-bearing dendritic cells (DCs), which are typically short-lived in lymphoid tissues. 'Survival gene' Bcl-xl is critical for the lifespan of DCs in vivo. Here, we showed that in vivo coadministration of Bcl-xl under control of the DC-specific promoter (CD11c-Bcl-xl) and TRP2hsp70 DNA prolonged T-cell stimulation by DCs and augmented TRP2-specific-IFN-gamma-producing CD8+ T-cell responses. Consistent with these findings, enhanced protection and significant therapeutic immunity to B16 melanoma was generated by this coimmunization strategy, which also augmented therapeutic immunity to GL-26 tumor. In this B16 melanoma model, results from animal experiments with depletion of immune cells indicate that CD8+ T cells and NK cells are important in the antitumor immunity induced by this coimmunization strategy. These observations suggest that 'survival gene' Bcl-xl potentiates the magnitude of antigen-specific-CD8+ T-cell responses and the efficacy of antitumor immunity induced by DNA vaccine, and is relevant for the design of in vivo targeted DC-based vaccine strategies to improve immunity against cancer.
Collapse
Affiliation(s)
- J H Kim
- Department of Dermatology and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Paschen A, Song M, Osen W, Nguyen XD, Mueller-Berghaus J, Fink D, Daniel N, Donzeau M, Nagel W, Kropshofer H, Schadendorf D. Detection of Spontaneous CD4+ T-Cell Responses in Melanoma Patients against a Tyrosinase-Related Protein-2–Derived Epitope Identified in HLA-DRB1*0301 Transgenic Mice. Clin Cancer Res 2005; 11:5241-7. [PMID: 16033842 DOI: 10.1158/1078-0432.ccr-05-0170] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The frequently expressed differentiation antigen tyrosinase-related protein-2 (TRP-2) has repeatedly been described as a target of spontaneous cytotoxic T-cell responses in melanoma patients, suggesting that it might be an ideal candidate antigen for T cell-based immunotherapy. As a prerequisite for immunization, T-cell epitopes have to be identified. Whereas a number of HLA class I-presented TRP-2-derived epitopes are known, information about HLA class II-presented antigenic ligands recognized by CD4+ T helper (Th) cells is limited. EXPERIMENTAL DESIGN The search for TRP-2-derived Th epitopes was carried out by competitive in vitro peptide binding studies with predicted HLA-DRB1*0301 ligands in combination with peptide and protein immunizations of HLA-DRB1*0301 transgenic mice. In vivo selected candidate epitopes were subsequently verified for their immunogenicity in human T-cell cultures. RESULTS This strategy led to the characterization of TRP-2(60-74) as an HLA-DRB1*0301-restricted Th epitope. Importantly, TRP-2(60-74)-reactive human CD4+ Th cell lines, specifically recognizing target cells loaded with recombinant TRP-2 protein, could be established by repeated peptide stimulation of peripheral blood lymphocytes from several HLA-DRB1*03+ melanoma patients. Even short-term peptide stimulation of patients' peripheral blood lymphocytes showed the presence of TRP-2(60-74)-reactive T cells, suggesting that these T cells were already activated in vivo. CONCLUSION Peptide TRP-2(60-74) might be a useful tool for the improvement of immunotherapy and immune monitoring of melanoma patients.
Collapse
Affiliation(s)
- Annette Paschen
- Skin Cancer Unit of the German Cancer Research Center Heidelberg, University Hospital Mannheim, Mannheim, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wlazlo AP, Deng H, Giles-Davis W, Ertl HCJ. DNA vaccines against the human papillomavirus type 16 E6 or E7 oncoproteins. Cancer Gene Ther 2004; 11:457-64. [PMID: 15118761 DOI: 10.1038/sj.cgt.7700723] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
DNA vaccines expressing the E6 or E7 oncoproteins of human papilloma virus type 16 (HPV-16) in either their wild-type form or fused to sequences that affect intracellular trafficking were tested for induction of protective immunity against tumor cell challenge in two models based on BALB/c and C57Bl/6 mice. The DNA vaccines to E7 gave uniformly disappointing results, while the DNA vaccine that expressed E6 linked to a viral leader sequence protected BALB/c mice against tumor cell challenge given before or after vaccination. The efficacy of this vaccine could be enhanced by a DNA vector prime/viral vector boost regimen. In contrast, priming of mice with the DNA vaccines to E7 reduced the efficacy of a viral vector expressing the same antigen.
Collapse
Affiliation(s)
- Anthony P Wlazlo
- The Wistar Institute, 3601 Spruce St., Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|