1
|
Ren AL, Wu JY, Lee SY, Lim M. Translational Models in Glioma Immunotherapy Research. Curr Oncol 2023; 30:5704-5718. [PMID: 37366911 DOI: 10.3390/curroncol30060428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023] Open
Abstract
Immunotherapy is a promising therapeutic domain for the treatment of gliomas. However, clinical trials of various immunotherapeutic modalities have not yielded significant improvements in patient survival. Preclinical models for glioma research should faithfully represent clinically observed features regarding glioma behavior, mutational load, tumor interactions with stromal cells, and immunosuppressive mechanisms. In this review, we dive into the common preclinical models used in glioma immunology, discuss their advantages and disadvantages, and highlight examples of their utilization in translational research.
Collapse
Affiliation(s)
- Alexander L Ren
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Janet Y Wu
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Si Yeon Lee
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| |
Collapse
|
2
|
Mercogliano MF, Bruni S, Mauro F, Elizalde PV, Schillaci R. Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13030564. [PMID: 33540543 PMCID: PMC7985780 DOI: 10.3390/cancers13030564] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/17/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine known to have contradictory roles in oncoimmunology. Indeed, TNFα has a central role in the onset of the immune response, inducing both activation and the effector function of macrophages, dendritic cells, natural killer (NK) cells, and B and T lymphocytes. Within the tumor microenvironment, however, TNFα is one of the main mediators of cancer-related inflammation. It is involved in the recruitment and differentiation of immune suppressor cells, leading to evasion of tumor immune surveillance. These characteristics turn TNFα into an attractive target to overcome therapy resistance and tackle cancer. This review focuses on the diverse molecular mechanisms that place TNFα as a source of resistance to immunotherapy such as monoclonal antibodies against cancer cells or immune checkpoints and adoptive cell therapy. We also expose the benefits of TNFα blocking strategies in combination with immunotherapy to improve the antitumor effect and prevent or treat adverse immune-related effects.
Collapse
Affiliation(s)
- María Florencia Mercogliano
- Laboratorio de Biofisicoquímica de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIBICEN-CONICET), Buenos Aires 1428, Argentina;
| | - Sofía Bruni
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Florencia Mauro
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Patricia Virginia Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Roxana Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
- Correspondence: ; Tel.: +54-11-4783-2869; Fax: +54-11-4786-2564
| |
Collapse
|
3
|
Sanchez VE, Lynes JP, Walbridge S, Wang X, Edwards NA, Nwankwo AK, Sur HP, Dominah GA, Obungu A, Adamstein N, Dagur PK, Maric D, Munasinghe J, Heiss JD, Nduom EK. GL261 luciferase-expressing cells elicit an anti-tumor immune response: an evaluation of murine glioma models. Sci Rep 2020; 10:11003. [PMID: 32620877 PMCID: PMC7335060 DOI: 10.1038/s41598-020-67411-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/03/2020] [Indexed: 11/13/2022] Open
Abstract
Preclinical models that reliably recapitulate the immunosuppressive properties of human gliomas are essential to assess immune-based therapies. GL261 murine glioma cells are widely used as a syngeneic animal model of glioma, however, it has become common practice to transfect these cells with luciferase for fluorescent tumor tracking. The aim of this study was to compare the survival of mice injected with fluorescent or non-fluorescent GL261 cells and characterize the differences in their tumor microenvironment. Mice were intracranially implanted with GL261, GL261 Red-FLuc or GL261-Luc2 cells at varying doses. Cytokine profiles were evaluated by proteome microarray and Kaplan–Meier survival analysis was used to determine survival differences. Median survival for mice implanted with 5 × 104 GL261 cells was 18 to 21 days. The GL261 Red-FLuc implanted mice cells did not reach median survival at any tumor dose. Mice injected with 3 × 105 GL261-Luc2 cells reached median survival at 23 days. However, median survival was significantly prolonged to 37 days in mice implanted with 5 × 104 GL261-Luc2 cells. Additionally, proteomic analyses revealed significantly elevated inflammatory cytokines in the supernatants of the GL261 Red-FLuc cells and GL261-Luc2 cells. Our data suggest that GL261 Red-FLuc and GL261-Luc2 murine models elicit an anti-tumor immune response by increasing pro-inflammatory modulators.
Collapse
Affiliation(s)
- Victoria E Sanchez
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - John P Lynes
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Stuart Walbridge
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Xiang Wang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nancy A Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Anthony K Nwankwo
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Hannah P Sur
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Gifty A Dominah
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Arnold Obungu
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Adamstein
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Pradeep K Dagur
- Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Dragan Maric
- Flow Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeeva Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Edjah K Nduom
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA. .,Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Room 3D-20, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Vannini E, Maltese F, Olimpico F, Fabbri A, Costa M, Caleo M, Baroncelli L. Progression of motor deficits in glioma-bearing mice: impact of CNF1 therapy at symptomatic stages. Oncotarget 2017; 8:23539-23550. [PMID: 28212563 PMCID: PMC5410325 DOI: 10.18632/oncotarget.15328] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 01/23/2017] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive type of brain tumor. In this context, animal models represent excellent tools for the early detection and longitudinal mapping of neuronal dysfunction, that are critical in the preclinical validation of new therapeutic strategies. In a mouse glioma model, we developed sensitive behavioral readouts that allow early recognizing and following neurological symptoms. We injected GL261 cells into the primary motor cortex of syngenic mice and we used a battery of behavioral tests to longitudinally monitor the dysfunction induced by tumor growth. Grip strength test revealed an early onset of functional deficit associated to the glioma growth, with a significant forelimb weakness appearing 9 days after tumor inoculation. A later deficit was observed in the rotarod and in the grid walk tasks. Using this model, we found reduced tumor growth and maintenance of behavioral functions following treatment with Cytotoxic Necrotizing Factor 1 (CNF1) at a symptomatic stage. Our data provide a detailed and precise examination of how tumor growth reverberates on the behavioral functions of glioma-bearing mice, providing normative data for the study of therapeutic strategies for glioma treatment. The reduced tumor volume and robust functional sparing observed in CNF1-treated, glioma-bearing mice strengthen the notion that CNF1 delivery is a promising strategy for glioma therapy.
Collapse
Affiliation(s)
- Eleonora Vannini
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Federica Maltese
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | | | | | - Mario Costa
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Matteo Caleo
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| |
Collapse
|
5
|
Immunocompetent murine models for the study of glioblastoma immunotherapy. J Transl Med 2014; 12:107. [PMID: 24779345 PMCID: PMC4012243 DOI: 10.1186/1479-5876-12-107] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/16/2014] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma remains a lethal diagnosis with a 5-year survival rate of less than 10%. (NEJM 352:987-96, 2005) Although immunotherapy-based approaches are capable of inducing detectable immune responses against tumor-specific antigens, improvements in clinical outcomes are modest, in no small part due to tumor-induced immunosuppressive mechanisms that promote immune escape and immuno-resistance. Immunotherapeutic strategies aimed at bolstering the immune response while neutralizing immunosuppression will play a critical role in improving treatment outcomes for glioblastoma patients. In vivo murine models of glioma provide an invaluable resource to achieving that end, and their use is an essential part of the preclinical workup for novel therapeutics that need to be tested in animal models prior to testing experimental therapies in patients. In this article, we review five contemporary immunocompetent mouse models, GL261 (C57BL/6), GL26 (C57BL/6) CT-2A (C57BL/6), SMA-560 (VM/Dk), and 4C8 (B6D2F1), each of which offer a suitable platform for testing novel immunotherapeutic approaches.
Collapse
|
6
|
Marshall D, Mitchell DA, Graner MW, Bigner DD. Immunotherapy of brain tumors. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:309-30. [PMID: 22230450 DOI: 10.1016/b978-0-444-52138-5.00020-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
7
|
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: 71] [Impact Index Per Article: 5.5] [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
|
8
|
Immunotherapy of brain cancers: the past, the present, and future directions. Clin Dev Immunol 2011; 2010:296453. [PMID: 21437175 PMCID: PMC3061456 DOI: 10.1155/2010/296453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Treatment of brain cancers, especially high grade gliomas (WHO stage III and IV) is slowly making progress, but not as fast as medical researchers and the patients would like. Immunotherapy offers the opportunity to allow the patient's own immune system a chance to help eliminate the cancer. Immunotherapy's strength is that it efficiently treats relatively small tumors in experimental animal models. For some patients, immunotherapy has worked for them while not showing long-term toxicity. In this paper, we will trace the history of immunotherapy for brain cancers. We will also highlight some of the possible directions that this field may be taking in the immediate future for improving this therapeutic option.
Collapse
|
9
|
Maes W, Van Gool SW. Experimental immunotherapy for malignant glioma: lessons from two decades of research in the GL261 model. Cancer Immunol Immunother 2011; 60:153-60. [PMID: 21120655 PMCID: PMC11028904 DOI: 10.1007/s00262-010-0946-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 11/05/2010] [Indexed: 02/04/2023]
Abstract
Nearly twenty years of experimental immunotherapy for malignant glioma yielded important insights in the mechanisms governing glioma immunology. Still considered promising, it is clear that immunotherapy does not on its own represent the magic bullet in glioma therapy. In this review, we summarize the major immunotherapeutic achievements in the mouse GL261 glioma model, which has emerged as the gold standard syngeneic model for experimental glioma therapy. Gene therapy, monoclonal antibody treatment, cytokine therapy, cell transfer strategies and dendritic cell therapy were hereby considered. Apart from the considerable progress made in understanding glioma immunology in this model, we also addressed its most pertinent issues and shortcomings. Despite these, the GL261 model will remain indispensable in glioma research since it is a fast, highly reproducible and easy-to-establish model system.
Collapse
Affiliation(s)
- Wim Maes
- Laboratory of Experimental Immunology (WM and SWVG), Pediatric Hemato-oncology (SWVG), University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Stefaan W. Van Gool
- Laboratory of Experimental Immunology (WM and SWVG), Pediatric Hemato-oncology (SWVG), University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
10
|
Thomas DL, Kim M, Bowerman NA, Narayanan S, Kranz DM, Schreiber H, Roy EJ. Recurrence of Intracranial Tumors following Adoptive T Cell Therapy Can Be Prevented by Direct and Indirect Killing Aided by High Levels of Tumor Antigen Cross-Presented on Stromal Cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:1828-37. [PMID: 19592642 DOI: 10.4049/jimmunol.0802322] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Elimination of peripheral tumors by adoptively transferred tumor-specific T cells may require killing of cancer cells and tumor stromal cells. Tumor Ags are cross-presented on stromal cells, resulting in direct cytotoxic T cell (CTL) killing of both Ag-expressing cancer cells and stromal cells. Indirect killing of Ag loss variant cells also occurs. We show here that similar processes occur in a brain tumor stromal environment. We used murine cancer cell lines that express high or low levels of a peptide Ag, SIYRYYGL (SIY), recognized by transgenic 2C CD8(+) T cells. The two cell lines are killed with equivalent efficiency by 2C T cells in vitro. Following adoptive transfer of 2C T cells into mice with established SIY-Hi or SIY-Lo brain tumors, tumors of both types regressed, but low-Ag-expressing tumors recurred. High-Ag-expressing tumors contained CD11b(+) cells cross-presenting SIY peptide and were completely eliminated by 2C T cells. To further test the role of cross-presentation, RAG1(-/-) H-2(b) mice were infused with H-2(k) tumor cells expressing high levels of SIY peptide. Adoptively transferred 2C T cells are able to kill cross-presenting H-2(b) stromal cells but not H-2(k) tumor cells. In peripheral models, this paradigm led to a small static tumor. In the brain, activated 2C T cells were able to kill cross-presenting CD11b(+) cells and completely eliminate the H-2(k) tumors in most mice. Targeting brain tumor stroma or increasing Ag shedding from tumor cells to enhance cross-presentation may improve the clinical success of T cell adoptive therapies.
Collapse
Affiliation(s)
- Diana L Thomas
- University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Accumulating evidence suggests that only a fraction of neoplastic cells, defined as cancer stem cells (CSC), are responsible for tumor perpetuation. Recent data suggest that neurospheres (NS) from glioblastoma multiforme (GBM) are enriched in CSC. The characterization of this subpopulation of brain tumor cells with a potent tumorigenic activity supports the cancer stem cell hypothesis in solid tumors and may imply that cancer cells are differentially targeted by treatments, including dendritic cell (DC) immunotherapy. To test therapeutic strategies, a good model mimicking the characteristics of GBM-NS and GBM-AC (Adherent Cells) was necessary. One of the most frequently used murine brain tumor models is the GL261 glioma cell line. To see whether GL261 cells could mimic the growth of human GBM-CSC we let them grow in EGF/bFGF without serum. After 5 days neurospheres were visible in the culture medium and were proliferating continuously. The characterization in vivo and in vitro demonstrates that GL261-NS satisfy criteria used to identify CSC and are more immunogenic than AC. DC loaded with GL261-NS lysates protect mice against tumors from both GL261-NS and GL261-AC. Our results suggest that only DC vaccination against neurospheres can restrain the growth of a highly infiltrating and aggressive model of glioma and may have implications for the design of novel, more effective immunotherapy trials for malignant glioma and possibly other malignancies.
Collapse
Affiliation(s)
- Serena Pellegatta
- Unit of Experimental Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Besta, Milano, Italy
| | | |
Collapse
|
12
|
Abstract
T-cell mediated immunotherapy is a conceptually attractive treatment option to envisage for glioma, since T lymphocytes can actively seek out neoplastic cells in the brain, and they have the potential to safely and specifically eliminate tumor. Some antigenic targets on glioma cells are already defined, and we can be optimistic that more will be discovered from progress in T-cell epitope identification and gene expression profiling of brain tumors. In parallel, advances in immunology (regional immunology, neuroimmunology, tumor immunology) now equip us to build upon the results from current immunotherapy trials in which the safety and feasibility of brain tumor immunotherapy have already been confirmed. We can now look to the next phase of immunotherapy, in which we must harness the most promising basic science advances and existing clinical expertise, and apply these to randomized clinical trials to determine the real clinical impact and applicability of these approaches for treating patients with currently incurable malignant brain tumors.
Collapse
Affiliation(s)
- Erwin G. Meir
- School of Medicine, Emory University, Clifton Road 1365C, Atlanta, 30322 U.S.A
| |
Collapse
|
13
|
Zheng R, Cohen PA, Paustian CA, Johnson TD, Lee WT, Shu S, Koski GK. Paired Toll-like receptor agonists enhance vaccine therapy through induction of interleukin-12. Cancer Res 2008; 68:4045-9. [PMID: 18519662 DOI: 10.1158/0008-5472.can-07-6669] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Minimal requirements for generating effective immunity include the delivery of antigenic (signal 1) and costimulatory (signal 2) signals to T lymphocytes. Recently, a class of third signals, often delivered by antigen-presenting dendritic cells, has been shown to greatly enhance immune responses, especially against tumors. Among signal 3 factors, interleukin (IL)-12 is particularly effective and can be conditionally induced by agonists of Toll-like transmembrane receptors (TLR). In this study, we assessed the therapeutic effect of adjuvant TLR agonist administration upon the capacity of dendritic cell (DC)-tumor electrofusion hybrids to eradicate established MCA205 sarcomas in syngeneic mice. Paired, but not solitary combinations of polyinosine:polycytadilic acid (P[I:C]; TLR3 agonist) and CpG DNA (ODN1826l; TLR9 agonist) stimulated IL-12 secretion from DCs in vitro and synergized with vaccination to achieve potent tumor rejection. Therapeutic effects, however, required coadministration of paired TLR agonists and DC-tumor fusion hybrids. The administration of TLR agonists alone or with fusion vaccine induced transient splenomegaly but without apparent toxicity. The therapeutic effects of this immunization regimen were significantly abrogated through the neutralization of IL-12p70, indicating that production of this third signal was essential to the observed tumor regression. These results show the profound functional consequences of TLR cooperativity and further highlight the critical role of IL-12 in antitumor immunity.
Collapse
Affiliation(s)
- Rongxiu Zheng
- Center for Surgery Research, Department of Immunology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Liu Z, Fan H, Wu Y, Chen B. Potent in vivo anti-tumor activity of isolated CD62L(low) lymph node cells sensitized in vivo with tumor lysate-pulsed DC-based vaccines. Cytotherapy 2008; 7:353-62. [PMID: 16162457 DOI: 10.1080/14653240500241925] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND DC are potent APC that can activate both CD4 and CD8 T cells in vitro and in vivo. Although the efficacy of DC-based cancer vaccines is currently being evaluated in clinical trials, the systemic immune suppression in cancer patients negatively impacts the clinical benefit of this therapeutic approach. Therefore, in this study we tested the feasibility and anti-tumor effect of adoptive immunotherapy using in vitro-activated CD62L(low) lymph node cells that were isolated from DC-vaccinated draining lymph nodes (VDLN). METHODS DC were prepared from BM cells and loaded with tumor lysate for inoculating into naive mice. Subsequently, the VDLN were removed and CD62L(low) cells in the VDLN population isolated, expanded in vitro by 5-day culture with IL-2 and immobilized anti-CD3 stimulation, then injected into mice with established pulmonary tumors. Eighteen days after treatment, mice were killed in order to enumerate pulmonary tumor nodes. RESULTS DC phagocytosed the tumor lysate efficiently and induced detectable T-cell responses and significant cell expansion in the draining lymph nodes. After induction of maturation by LPS treatment, DC expressed higher levels of CD40, CD86 and MHC class II molecules. When CD62L(low) VDLN cells that had been isolated and expanded in vitro were transferred into tumor-bearing mice, as few as 3 x 10(6) cells were able to cure metastatic pulmonary tumors in vivo. DISCUSSION DC-based VDLN T cells are an important source of anti-tumor effector for adoptive immunotherapy. This study provides a novel and an effective protocol using T-cell adoptive immunotherapy for application in cancer patients; therefore, clinical trials based on this protocol may be warranted.
Collapse
Affiliation(s)
- Z Liu
- Department of Thoracic Surgery, Tongji University Affiliated East Hospital, China
| | | | | | | |
Collapse
|
15
|
Ishida A, Tanaka H, Hiura T, Miura S, Watanabe S, Matsuyama K, Kuriyama H, Tanaka J, Kagamu H, Gejyo F, Yoshizawa H. Generation of anti-tumour effector T cells from naïve T cells by stimulation with dendritic/tumour fusion cells. Scand J Immunol 2007; 66:546-54. [PMID: 17953530 DOI: 10.1111/j.1365-3083.2007.02012.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumour-draining lymph node T cells are an excellent source of effector T cells that can be used in adoptive tumour immunotherapy because they have already been sensitized to tumour-associated antigens in vivo. However, such tumour-specific immune cells are not readily obtained from the host due to poor immunogenicity of tumours and reduced host immune responses. One obstacle in implementation of adoptive immunotherapy has been insufficient sensitization and expansion of tumour-specific effector cells. In this study, we aim to improve adoptive immunotherapy by generating anti-tumour effector T cells from naïve T lymphocytes. We attempted to achieve this by harnessing the advantages of dendritic cell (DC)-based anti-cancer vaccine strategies. Electrofusion was routinely employed to produce fusion cells with 30-40% efficiency by using the poorly immunogenic murine B16/F10 cell line, D5 cells, and DC generated from bone marrow cells. CD62L-positive T cells from spleens of naïve mice and the fusion cells were cocultured with a low concentration of IL-2. After 9 days of culture, the antigen-specific T cells were identified with an upregulation of CD25 and CD69 expression and a downregulation of CD62L expression. These cells secreted IFN-gamma upon stimulation with irradiated tumour cells. Moreover, when transferred into mice with 3-day established pulmonary metastases, these cells with coadministration of IL-2 exhibited anti-tumour efficacy. In contrast, naïve T cells cocultured with a mixture of unfused DC and irradiated tumour cells did not exhibit anti-tumour efficacy. Our strategy provides the basis for a new approach in adoptive T cell immunotherapy for cancer.
Collapse
Affiliation(s)
- A Ishida
- Division of Respiratory Medicine, Department of Homeostatic Regulation and Development, Course for Biological Functions and Medical Control, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ehtesham M, Black KL, Yu JS. Recent progress in immunotherapy for malignant glioma: treatment strategies and results from clinical trials. Cancer Control 2007; 11:192-207. [PMID: 15153843 DOI: 10.1177/107327480401100307] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Despite advances in surgical and adjuvant radiation therapy and chemotherapy strategies, malignant gliomas continue to be associated with poor prognoses. METHODS We review immune-mediated treatment approaches for malignant glioma and the relevance of recent clinical trials and their outcomes. We specifically address the increasing evidence implicating the role of cytotoxic T cells in ensuring adequate immune-mediated clearance of neoplastic cells and the need for the optimization of therapies that can elicit and support such antitumor T-cell activity. RESULTS The poor outcome of this disease has spurred the search for novel experimental therapies that can address and overcome the root biological phenomena associated with the lethality of this disease. The use of immunotherapy to bolster the otherwise impaired antitumor immune responses in glioma patients has received increasing attention. CONCLUSIONS An effective treatment paradigm for malignant gliomas may eventually require a multifaceted approach combining two or more different immunotherapeutic strategies. Such scenarios may involve the use of local cytokine gene therapy to enhance glioma-cell immunogenicity in conjunction with dendritic cell-based active vaccination to stimulate systemic tumoricidal T-cell immunity. Given the heterogeneity of this disease process and the potential risk of immunoediting out a selected, treatment-refractory tumor cell population, the concurrent use of multiple modalities that target disparate tumor characteristics may be of greatest therapeutic relevance.
Collapse
Affiliation(s)
- Moneeb Ehtesham
- Maxine Dunitz Neurosurgical, Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
17
|
Ye Z, Shi M, Chan T, Sas S, Xu S, Xiang J. Engineered CD8+ cytotoxic T cells with fiber-modified adenovirus-mediated TNF-alpha gene transfection counteract immunosuppressive interleukin-10-secreting lung metastasis and solid tumors. Cancer Gene Ther 2007; 14:661-75. [PMID: 17479109 DOI: 10.1038/sj.cgt.7701039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
T-cell suppression derived from tumor-secreted immunosuppressive interleukin (IL)-10 becomes a major barrier to CD8+ T-cell immunotherapy of tumors. Tumor necrosis factor-alpha (TNF-alpha) is a multifunctional cytokine capable of activating T and dendritic cells (DCs) and counteracting IL-10-mediated DC inhibition and regulatory T-cell-mediated immune suppression. In this study, we constructed a recombinant adenovirus (MF)AdVTNF with fiber-gene modified by RGD insertion into the viral knob's H1 loop and a melanoma cell line B16(OVA/IL-10) engineered to express ovalbumin (OVA) and to secrete IL-10 (2.2 ng/ml/10(6) cells/24 h). We transfected OVA-specific CD8+ T cells with (MF)AdVTNF, and found a fivefold increase in transgene human TNF-alpha secretion (4.3 ng/ml/10(6) cells/24 h) by the engineered CD8+ T(TNF) cells transfected with (MF)AdVTNF, compared to that (0.8 ng/ml/10(6) cells/24 h) by CD8+ T cells transfected with the original AdVTNF without viral fiber modification. The engineered CD8+ T(TNF) cells exhibited enhanced cytotoxicity and elongated survival in vivo after adoptive transfer. TNF-alpha derived from both the donor CD8+ T cells and the host cells plays an important role in donor CD8+ T-cell survival in vivo after adoptive transfer. We also demonstrated that the transfected B16(OVA/IL-10) tumor cells secreting IL-10 are more resistant to in vivo CD8+ T-cell therapy than the original B16(OVA) tumor cells without IL-10 expression. Interestingly, the engineered CD8+ T(TNF) cells secreting transgene-coded TNF-alpha, but not the control CD8+ T(control) cells without any transgene expression eradicated IL-10-secreting 12-day lung micrometastasis in all 10/10 mice and IL-10-secreting solid tumors ( approximately 5 mm in diameter) in 6/10 mice. Transfer of the engineered CD8+ T(TNF) cells further induced both donor- and host-derived memory CD8+ T cells, leading to a stronger long-term antitumor immunity against the IL-10-secreting B16(OVA/IL-10) tumor cell challenges. Therefore, CD8+ T cells engineered to secrete TNF-alpha may be useful when designing strategies for adoptive T-cell therapy of solid tumors.
Collapse
Affiliation(s)
- Z Ye
- Research Unit, Health Research Division, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, Canada
| | | | | | | | | | | |
Collapse
|
18
|
Ryan CM, Schell TD. Accumulation of CD8+T Cells in Advanced-Stage Tumors and Delay of Disease Progression following Secondary Immunization against an Immunorecessive Epitope. THE JOURNAL OF IMMUNOLOGY 2006; 177:255-67. [PMID: 16785521 DOI: 10.4049/jimmunol.177.1.255] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Self-reactive T cells that survive the process of positive and negative selection during thymocyte development represent potential effector cells against tumors that express these same self-Ags. We have previously shown that CD8+ T lymphocytes (T(CD8)) specific for an immunorecessive epitope, designated epitope V, from the SV40 large T Ag (Tag) escape thymic deletion in line SV11 Tag-transgenic mice. In contrast, these mice are tolerant to the three most dominant Tag epitopes. The majority of the residual epitope V-specific T(CD8) have a low avidity for the target epitope, but a prime/boost regimen can expand higher avidity clones in vivo. Whether higher avidity T(CD8) targeting this epitope are affected by Tag-expressing tumors in the periphery or can be recruited for control of tumor progression remains unknown. In the current study, we determined the fate of naive TCR-transgenic T(CD8) specific for Tag epitope V (TCR-V cells) following transfer into SV11 mice bearing advanced-stage choroid plexus tumors. The results indicate that TCR-V cells are rapidly triggered by the endogenous Tag and acquire effector function, but fail to accumulate within the tumors. Primary immunization enhanced TCR-V cell frequency in the periphery and promoted entry into the brain, but a subsequent booster immunization caused a dramatic accumulation of TCR-V T cells within the tumors and inhibited tumor progression. These results indicate that epitope V provides a target for CD8+ T cells against spontaneous tumors in vivo, and suggests that epitopes with similar properties can be harnessed for tumor immunotherapy.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Polyomavirus Transforming/administration & dosage
- Antigens, Polyomavirus Transforming/biosynthesis
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/immunology
- Brain Neoplasms/immunology
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Brain Neoplasms/prevention & control
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/transplantation
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Movement/immunology
- Choroid Plexus/immunology
- Choroid Plexus/pathology
- Cytotoxicity Tests, Immunologic
- Disease Progression
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Genes, Recessive/immunology
- Immunization, Secondary/methods
- Lymphocyte Activation/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Staging
- Polyomavirus Infections/immunology
- Polyomavirus Infections/mortality
- Polyomavirus Infections/pathology
- Polyomavirus Infections/prevention & control
- Protein Structure, Tertiary/genetics
- Receptors, Antigen, T-Cell/genetics
- Survival Analysis
- Tumor Virus Infections/immunology
- Tumor Virus Infections/mortality
- Tumor Virus Infections/pathology
- Tumor Virus Infections/prevention & control
Collapse
Affiliation(s)
- Christina M Ryan
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | |
Collapse
|
19
|
Szatmári T, Lumniczky K, Désaknai S, Trajcevski S, Hídvégi EJ, Hamada H, Sáfrány G. Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci 2006; 97:546-53. [PMID: 16734735 PMCID: PMC11159227 DOI: 10.1111/j.1349-7006.2006.00208.x] [Citation(s) in RCA: 226] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Mouse glioma 261 (Gl261) cells are used frequently in experimental glioblastoma therapy; however, no detailed description of the Gl261 tumor model is available. Here we present that Gl261 cells carry point mutations in the K-ras and p53 genes. Basal major histocompatibility complex (MHC)I, but not MHCII, expression was detected in Gl261 cells. The introduction of interferon-gamma-encoding genes increased expression of both MHCI and MHCII. A low amount of B7-1 and B7-2 RNA was detected in wild-type cells, but cytokine production did not change expression levels. Gl261 cells were transduced efficiently by adenoviral vectors; the infectivity of retroviral vectors was limited. Low numbers of transplanted Gl261 cells formed both subcutaneous and intracranial tumors in C57BL/6 mice. The cells were moderately immunogenic: prevaccination of mice with irradiated tumor cells 7 days before intracranial tumor challenge prevented tumor formation in approximately 90% of mice. When vaccination was carried out on the day or 3 days after tumor challenge, no surviving animals could be found. In vitro-growing cells were radiosensitive: less than 2 Gy was required to achieve 50% cell mortality. Local tumor irradiation with 4 Gy X-rays in brain tumor-bearing mice slowed down tumor progression, but none of the mice were cured off the tumor. In conclusion, the Gl261 brain tumor model might be efficiently used to study the antitumor effects of various therapeutic modalities, but the moderate immunogenicity of the cells should be considered.
Collapse
Affiliation(s)
- Tünde Szatmári
- Department of Molecular and Tumor Radiobiology, Frederic Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest 1221, Hungary
| | | | | | | | | | | | | |
Collapse
|
20
|
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: 1.2] [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
|
21
|
Prins RM, Craft N, Bruhn KW, Khan-Farooqi H, Koya RC, Stripecke R, Miller JF, Liau LM. The TLR-7 agonist, imiquimod, enhances dendritic cell survival and promotes tumor antigen-specific T cell priming: relation to central nervous system antitumor immunity. THE JOURNAL OF IMMUNOLOGY 2006; 176:157-64. [PMID: 16365406 DOI: 10.4049/jimmunol.176.1.157] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunotherapy represents an appealing option to specifically target CNS tumors using the immune system. In this report, we tested whether adjunctive treatment with the TLR-7 agonist imiquimod could augment antitumor immune responsiveness in CNS tumor-bearing mice treated with human gp100 + tyrosine-related protein-2 melanoma-associated Ag peptide-pulsed dendritic cell (DC) vaccination. Treatment of mice with 5% imiquimod resulted in synergistic reduction in CNS tumor growth compared with melanoma-associated Ag-pulsed DC vaccination alone. Continuous imiquimod administration in CNS tumor-bearing mice, however, was associated with the appearance of robust innate immune cell infiltration and hemorrhage into the brain and the tumor. To understand the immunological mechanisms by which imiquimod augmented antitumor immunity, we tested whether imiquimod treatment enhanced DC function or the priming of tumor-specific CD8+ T cells in vivo. With bioluminescent, in vivo imaging, we determined that imiquimod dramatically enhanced both the persistence and trafficking of DCs into the draining lymph nodes after vaccination. We additionally demonstrated that imiquimod administration significantly increased the accumulation of tumor-specific CD8+ T cells in the spleen and draining lymph nodes after DC vaccination. The results suggest that imiquimod positively influences DC trafficking and the priming of tumor-specific CD8+ T cells. However, inflammatory responses induced in the brain by TLR signaling must also take into account the local microenvironment in the context of antitumor immunity to induce clinical benefit. Nevertheless, immunotherapeutic targeting of malignant CNS tumors may be enhanced by the administration of the innate immune response modifier imiquimod.
Collapse
Affiliation(s)
- Robert M Prins
- Division of Neurosurgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
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: 0.9] [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
|
23
|
Huang H, Bi XG, Yuan JY, Xu SL, Guo XL, Xiang J. Combined CD4+ Th1 effect and lymphotactin transgene expression enhance CD8+ Tc1 tumor localization and therapy. Gene Ther 2005; 12:999-1010. [PMID: 15789061 DOI: 10.1038/sj.gt.3302486] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Type 1 T cells are the major components in antitumor immunity. The lack of efficient CD8(+) cytotoxic T (Tc) cell infiltration of tumors is a major obstacle to adoptive Tc-cell therapy. We have previously demonstrated that adenovirus (AdV)-mediated transgene lymphotactin (Lptn) expression by intratumoral AdVLptn injection and intravenous CD4(+) helper T (Th) cell transfer can enhance Tc-cell tumor infiltration and eradication of early stage tumors (5 mm in diameter). In this study, we generated ovalbumin (OVA)-specific Tc1 and Th1 cells in vitro by incubation of OVA-pulsed dendritic cells with naive T cells from T-cell receptor (TCR) transgenic OT I and OT II mice. We then investigated the potential synergy of Th1 help effect and Lptn transgene expression in Tc1-cell therapy of well-established OVA-expressing EG7 solid tumors (7 mm in diameter). Our data showed that a combined adoptive T-cell therapy of Th1 (2.5 x 10(6) cells per mouse) and Tc1 (5 x 10(6) cells per mouse) resulted in regression of all eight (100%) transgene Lptn expressed EG7 tumors, which is significantly higher than four from eight (50%) in AdVLptn/Tc1 group and two from eight (25%) in Tc1/Th1 group (P < 0.05). The amount of transferred Tc1 cells detected in Lptn-expressed tumors with Th1 treatment is 0.72%, which is significantly higher than those of AdVLptn (0.22%), Th1 (0.41%) and the control AdVpLpA (0.09%) treatment groups (P < 0.05). Enhanced Tc1 tumor localization may be derived from the chemotactic effect of Lptn and the proliferative effect of Th1 and Lptn. This novel therapeutic strategy with enhancement of Tc1 tumor localization in the therapy of well-established tumors may become a tool of considerable conceptual interest in the implementation of future clinical objectives.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adoptive Transfer/methods
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Cell Proliferation
- Chemokines, C
- Chemotaxis, Leukocyte
- Female
- Gene Expression
- Genetic Therapy/methods
- Genetic Vectors/administration & dosage
- Lymphocytes/immunology
- Lymphocytes, Tumor-Infiltrating
- Lymphokines/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Models, Animal
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Ovalbumin/immunology
- Receptors, Antigen, T-Cell/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sialoglycoproteins/genetics
- Th1 Cells/immunology
- Transduction, Genetic/methods
- Transgenes
Collapse
Affiliation(s)
- H Huang
- Department of Oncology, Research Unit, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, Canada
| | | | | | | | | | | |
Collapse
|
24
|
Kjaergaard J, Wang LX, Kuriyama H, Shu S, Plautz GE. Active immunotherapy for advanced intracranial murine tumors by using dendritic cell-tumor cell fusion vaccines. J Neurosurg 2005; 103:156-64. [PMID: 16121986 DOI: 10.3171/jns.2005.103.1.0156] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECT Immunotherapy for malignant brain tumors by active immunization or adoptive transfer of tumor antigen-specific T lymphocytes has the potential to make up for some of the limitations of current clinical therapy. In this study, the authors tested whether active immunotherapy is curative in mice bearing advanced, rapidly progressive intracranial tumors. METHODS Tumor vaccines were created through electrofusion of dendritic cells (DCs) and irradiated tumor cells to form multinucleated heterokaryons that retained the potent antigen processing and costimulatory function of DCs as well as the entire complement of tumor antigens. Murine hosts bearing intracranial GL261 glioma or MCA 205 fibrosarcoma were treated with a combination of local cranial radiotherapy, intrasplenic vaccination with DC/tumor fusion cells, and anti-OX40R (CD134) monoclonal antibody (mAb) 7 days after tumor inoculation. Whereas control mice had a median survival of approximately 20 days, the treated mice underwent complete tumor regression that was immunologically specific. Seven days after vaccination treated mice demonstrated robust infiltration of CD4+ and CD8+ T cells, which was exclusively confined to the tumor without apparent neurological toxicity. Cured mice survived longer than 120 days with no evidence of tumor recurrence and resisted intracranial tumor challenge. CONCLUSIONS These data indicate a strategy to achieve an antitumor response against tumors in the central nervous system that is highly focused from both immunological and anatomical perspectives.
Collapse
Affiliation(s)
- Jorgen Kjaergaard
- Center for Surgery Research, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
25
|
Wang LX, Huang WX, Graor H, Cohen PA, Kim JA, Shu S, Plautz GE. Adoptive immunotherapy of cancer with polyclonal, 108-fold hyperexpanded, CD4+ and CD8+ T cells. J Transl Med 2004; 2:41. [PMID: 15566571 PMCID: PMC535812 DOI: 10.1186/1479-5876-2-41] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2004] [Accepted: 11/26/2004] [Indexed: 12/05/2022] Open
Abstract
T cell-mediated cancer immunotherapy is dose dependent and optimally requires participation of antigen-specific CD4+ and CD8+ T cells. Here, we isolated tumor-sensitized T cells and activated them in vitro using conditions that led to greater than 108-fold numerical hyperexpansion of either the CD4+ or CD8+ subset while retaining their capacity for in vivo therapeutic efficacy. Murine tumor-draining lymph node (TDLN) cells were segregated to purify the CD62Llow subset, or the CD4+ subset thereof. Cells were then propagated through multiple cycles of anti-CD3 activation with IL-2 + IL-7 for the CD8+ subset, or IL-7 + IL-23 for the CD4+ subset. A broad repertoire of TCR Vbeta families was maintained throughout hyperexpansion, which was similar to the starting population. Adoptive transfer of hyper-expanded CD8+ T cells eliminated established pulmonary metastases, in an immunologically specific fashion without the requirement for adjunct IL-2. Hyper-expanded CD4+ T cells cured established tumors in intracranial or subcutaneous sites that were not susceptible to CD8+ T cells alone. Because accessibility and antigen presentation within metastases varies according to anatomic site, maintenance of a broad repertoire of both CD4+ and CD8+ T effector cells will augment the overall systemic efficacy of adoptive immunotherapy.
Collapse
Affiliation(s)
- Li-Xin Wang
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Wen-Xin Huang
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Hallie Graor
- Dept. of General Surgery, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Peter A Cohen
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Julian A Kim
- Dept. of General Surgery, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Suyu Shu
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Gregory E Plautz
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
26
|
Wang LX, Kjaergaard J, Cohen PA, Shu S, Plautz GE. Memory T cells originate from adoptively transferred effectors and reconstituting host cells after sequential lymphodepletion and adoptive immunotherapy. THE JOURNAL OF IMMUNOLOGY 2004; 172:3462-8. [PMID: 15004146 DOI: 10.4049/jimmunol.172.6.3462] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Adoptive transfer of tumor-specific effector T cells induces regression of advanced tumors and induces a long term memory response; however, the origin of this response has not been clearly defined. In this study Thy1.2+ mice bearing advanced MCA-205 tumors were treated with sublethal total body irradiation, followed by adoptive transfer of congenic Thy1.1+ T cells that had been sensitized to tumor in vivo and then activated ex vivo with anti-CD3, IL-2, and IL-7. Splenocytes were recovered >140 days after the initial therapy, and the L-selectinlow memory cell subset was separated into host Thy1.2+ and transferred Thy1.1+ cells and restimulated ex vivo. Both adoptively transferred Thy1.1+ cells as well as reconstituted host Thy1.2+ cells could specifically eliminate MCA-205 pulmonary metastases. Interestingly, hosts with partial responses followed by tumor recurrence nevertheless harbored memory cells that could be isolated and numerically amplified ex vivo to regenerate potent effector function. Memory cells were recovered after adoptive transfer into lymphodepleted nontumor-bearing hosts, indicating that they were not dependent on continued Ag exposure. These experiments establish that rapid ex vivo expansion of tumor Ag-primed T cells does not abrogate their capacity to become long-lived memory cells. Moreover, immune-mediated tumor regression coincident with lymphoid reconstitution produces another wave of host memory cells. These data suggest an approach to rescuing antitumor immune function even in hosts with long-standing progressive tumor through restorative ex vivo activation.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- Cell Differentiation/immunology
- Cell Line, Tumor
- Cell Survival/immunology
- Female
- Immunologic Memory
- Immunotherapy, Adoptive/methods
- Injections, Subcutaneous
- Lymphocyte Depletion/methods
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation/pathology
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/pathology
- Sarcoma, Experimental/prevention & control
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- T-Lymphocytes, Regulatory/transplantation
- Thy-1 Antigens/biosynthesis
Collapse
Affiliation(s)
- Li-Xin Wang
- Center for Surgery Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
27
|
Huang H, Xiang J. Synergistic effect of lymphotactin and interferon gamma-inducible protein-10 transgene expression in T-cell localization and adoptive T-cell therapy of tumors. Int J Cancer 2004; 109:817-25. [PMID: 15027114 DOI: 10.1002/ijc.20043] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The lack of efficient T-cell infiltration of tumors is a major obstacle to successful adoptive T-cell therapy. We have previously demonstrated that adenovirus (AdV)-mediated transgene lymphotactin (Lptn) or IP-10 expression in tumors can significantly enhance T-cell tumor infiltration. In this study, active OVA-specific CD8+ T cells were prepared by coculturing naive OVA-specific CD8+ T cells from transgenic OT I mice with OVA-I peptide-pulsed dendritic cells in vitro. These XCR-1- and CXCR3-expressing T cells predominantly secreted IFN-gamma and displayed significant killing activity (84% at effector:target cell ratio of 1.5) against OVA-expressing EG7 tumor cells through perforin-mediated pathway. Our data also showed that chemokine Lptn and IP-10 not only can chemoattract, but also stimulate proliferation of CD8+ T cells in vitro, and that a mixture of Lptn and IP-10 can more efficiently chemoattract CD8+ T cells than either one of them. Furthermore, we demonstrated that the transferred CD8+ T cells detected in group of tumors treated with both AdVLptn and AdVIP-10 (group a) are around 4 and 2 times more than that in groups of tumors treated with control AdVpLpA (group b) and either AdVIP-10 (group c) or AdVLptn (group d), respectively. Around 87.5% of mice in group a were tumor-free compared to the aggressive tumor growth in all 8 mice of group b and 25% or 37.5% cured mice seen in groups c and d (p<0.05). Thus, our results indicate that enhancement of adoptive T-cell therapy can be obtained by double tranmsgene Lptn and IP-10 expression, which facilitates CD8+ T-cell tumor localization through proliferation and chemoattraction of the transferred CD8+ T cells by in situ chemokine transgene expressions in the tumors. Collectively, our data provide solid evidence of a potent synergy between adoptive T-cell therapy and adenovirus-mediated Lptn and IP-10 gene transfer into tumor tissues, which culminated in the T-cell tumor localization and eradication of well-established tumor masses.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cell Division
- Chemokine CXCL10
- Chemokines, C
- Chemokines, CXC/genetics
- Chemotaxis, Leukocyte/immunology
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Drug Synergism
- Female
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Immunotherapy, Adoptive
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Lymphokines/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Ovalbumin/immunology
- Receptors, Antigen, T-Cell/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sialoglycoproteins/genetics
- Transgenes
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Hui Huang
- Research Unit, Saskatchewan Cancer Agency, Department of Microbiology, University of Saskatchewan, Saskatoon, Canada
| | | |
Collapse
|
28
|
Ali S, Ahmad M, Lynam J, Rees RC, Brown N. Trafficking of tumor peptide-specific cytotoxic T lymphocytes into the tumor microcirculation. Int J Cancer 2004; 110:239-44. [PMID: 15069688 DOI: 10.1002/ijc.20113] [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: 11/12/2022]
Abstract
The major histocompatibility complex class I-restricted CD8(+) cytotoxic T-lymphocyte (CTL) effector arm of the adaptive immune response can specifically recognize and destroy tumor cells expressing peptide antigens. Although adoptive T-cell therapy has been successfully used for the treatment of viral and malignant diseases, little is known of the trafficking and fate of adoptively transferred antigen-specific T cells. In the present study, splenocytes derived from mice that rejected their tumors (CT26 or CT26-clone 25 tumors) in response to direct intratumor injection of disabled infectious single-cycle herpes simplex virus (DISC-HSV) encoding murine GM-CSF were restimulated with peptide in vitro. CTLs specific for the AH-1 and beta-gal peptides expressed by CT26 and CT26-clone 25 tumor cells, respectively, were generated and used for adoptive cellular therapy and trafficking studies. Intravenous administration of AH-1-specific CTLs 3 days following i.v. injection of CT26 cells resulted in significant tumor growth inhibition, whereas administration of control CTLs generated against a bacterial beta-gal peptide did not inhibit the growth of tumors. Trafficking of AH-1-specific lymphocytes and their interaction with the CT26 tumor microcirculation was analyzed using real-time in vivo microscopy (IVM). AH-1-specific but not beta-gal-specific CTLs adhered and localized in the CT26 tumor microvasculature, but neither population adhered to the endothelium of the normal microcirculation. This study provides direct visual evidence suggesting that AH-1-specific CTLs that mediate a therapeutic response traffic to and localize within the tumor microenvironment.
Collapse
Affiliation(s)
- Selman Ali
- Interdisciplinary Biomedical Research Centre, School of Science, Nottingham Trent University, Nottingham, United Kingdom
| | | | | | | | | |
Collapse
|
29
|
Chan B, Lee W, Hu CXL, Ng P, Li KW, Lo G, Ho G, Yeung DW, Woo D. Adoptive cellular immunotherapy for non-small cell lung cancer: a pilot study. Cytotherapy 2003; 5:46-54. [PMID: 12745587 DOI: 10.1080/14653240310000074] [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: 10/27/2022]
Abstract
BACKGROUND Priming with autologous tumor vaccine followed by ex vivo expansion of activated T cells is a feasible experimental strategy. This paper describes the application of this cellular therapy to treat patients with late-stage non-small-cell lung cancer (NSCLC). METHODS Twenty-one patients with Stages III and IV NSCLC were treated. Tumor samples were obtained surgically (five patients) or by using aco-axial biopsy needle (16 patients). Each course of vaccination consisted of irradiated tumor cells, mixed with GM-CSF and injected intradermally on Day 1, followed by GM-CSF only on Days 2- 5. The course was repeated 10-14 days later. Lymphocytes were collected 10-14 day after the second course and ex vivo expanded using IL-2 and anti-CD3 Ab. The expansion products were then re-infused into the patients. RESULTS Twelve out of 16 biopsies resulted in optimal cell numbers for vaccine preparation. Nineteen out of 21 patients achieved a delayed type hypersensitivity (DTH response after two courses of vaccination. In 18/21 patients, the ex vivo expansion products contained > 1.6 x 10(10) cells. Subset analysis showed 77.0-97.2% T cells with a CD4:CD8 ratio of 0.65-4.0; natural killer cells were 2.0-18.6%. There were no significant toxicities. The median survival of all 21 patients was 18.6 months, with a 1-year survival of 51.6%. CONCLUSION Autologous tumor cell-vaccination may be combined with ex vivo expansion of lymphocytes as adoptive cellular immunotherapy for advanced NSCLC. Overall survival in this cohort of poor prognosis patients compared favorably with results reported in the literature.
Collapse
Affiliation(s)
- B Chan
- Biological Therapy Unit, Hong Kong Sanatorium & Hospital, Hong Kong, PRC
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Despite remarkable advancements in imaging modalities and treatment options available to patients diagnosed with malignant brain tumors, the prognosis for those with high-grade lesions remains poor. The imprecise mechanisms of currently available treatments to manage these tumors do not spare damage to the normal surrounding brain and often result in major cognitive and motor deficits. Immunotherapy holds the promise of offering a potent, yet targeted, treatment to patients with brain tumors, with the potential to eradicate the malignant tumor cells without damaging normal tissues. The T cells of the immune system are uniquely capable of recognizing the altered protein expression patterns within tumor cells and mediating their destruction through a variety of effector mechanisms. Adoptive T-cell therapy is an attempt to harness and amplify the tumor-eradicating capacity of a patients' own T cells and then return these effectors to the patient in such a state that they effectively eliminate residual tumor. Although this approach is not new to the field of tumor immunology, new advancements in our understanding of T-cell activation and function and breakthroughs in tumor antigen discovery hold great promise for the translation of this modality into a clinical success.
Collapse
Affiliation(s)
- Duane A Mitchell
- Department of Pathology and Department of Surgery, Duke University Medical Center, Box 3807, Durham, NC 27710, USA
| | | | | |
Collapse
|
31
|
Huang H, Liu Y, Xiang J. Synergistic effect of adoptive T-cell therapy and intratumoral interferon gamma-inducible protein-10 transgene expression in treatment of established tumors. Cell Immunol 2002; 217:12-22. [PMID: 12425997 DOI: 10.1016/s0008-8749(02)00508-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The lack of efficient T-cell infiltration of tumors is a major obstacle to successful adoptive T-cell therapy. We have previously shown that transplanted SP2/0 myeloma tumors engineered to express lymphotactin invariably induced tumor regress mediated by SP2/0 tumor-specific T cells. Herein, we further systemically characterize these activated T cells and investigate their therapeutic efficacy, either alone or with the chemokine interferon gamma (IFN-gamma)-inducible protein-10 (IP-10) gene therapy. Following stimulation with SP2/0 cells, these activated T cells were CD25(+)FasL(+) L-selectin(low), expressed CXCR3 receptor and were chemoattracted by IP-10 in vitro. They comprised 64% CD4(+) Th1 and 36% CD8(+) Tc1 cells, both of which expressed IFN-gamma, perforin, and TNF-alpha, but not IL-4. The activated T cells were strongly cytotoxic for SP2/0 tumor cells (79% specific killing; E:T ratio, 50), mainly via perforin-mediated pathway. Cell tracking using labeled T cells confirmed that these T cells infiltrated better into the IP-10-expressing tumors than non-IP-10-expressing ones. In vivo, combined intratumoral IP-10 gene transfer and adoptive T-cell immunotherapy for well-established SP2/0 tumors eradicated the tumors in 7 of the 8 mice. Control or IP-10 adenoviral treatments by themselves neither alter the lethal outcome for tumor-bearing mice nor did T-cell therapy by itself, although the latter two treatments did slow its time-frame. Taken together, our data provide solid evidence of a potent synergy between adoptive T-cell therapy and IP-10 gene transfer into tumor tissues, which culminated in the eradication of well-established tumor masses.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cell Movement
- Cells, Cultured
- Chemokine CXCL10
- Chemokines, C
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Combined Modality Therapy
- Genetic Therapy
- Genetic Vectors
- Immunotherapy, Adoptive
- Lymphocyte Activation
- Lymphokines/genetics
- Mice
- Mice, Inbred BALB C
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Multiple Myeloma/therapy
- Neoplasm Transplantation
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Neovascularization, Pathologic
- Sialoglycoproteins/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- Th1 Cells/immunology
- Transgenes
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Hui Huang
- Research Unit, Saskatchewan Cancer Agency, Department of Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 4H4
| | | | | |
Collapse
|
32
|
Carlos TM. Leukocyte recruitment at sites of tumor: dissonant orchestration. J Leukoc Biol 2001. [DOI: 10.1189/jlb.70.2.171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- T. M. Carlos
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
33
|
Radoja S, Saio M, Frey AB. CD8+ tumor-infiltrating lymphocytes are primed for Fas-mediated activation-induced cell death but are not apoptotic in situ. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:6074-83. [PMID: 11342625 DOI: 10.4049/jimmunol.166.10.6074] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Induction of Fas-mediated activation-induced cell death in antitumor T cells has been hypothesized to permit tumor escape from immune destruction. Several laboratories have proposed that expression of Fas ligand (L) by tumor is the basis for this form of T cell tolerance. In this study, we characterized murine tumor-infiltrating lymphocytes (TIL) for activation status, cell cycle status, level of apoptosis, cytokine secretion, and proliferative capacity. TILs express multiple activation markers (circa CD69, CD95L, CD122, and LFA-1) and contain IL-2 and IFN-gamma mRNAs, but are neither cycling nor apoptotic in situ. In addition, TIL are dramatically suppressed in proliferative response and do not secrete IL-2 and IFN-gamma. However, upon purification and activation in vitro, TIL secrete high levels of IL-2 and IFN-gamma, enter S phase, and then die by Fas-mediated apoptosis. Activation by injection of anti-TCR Ab or IL-2 into tumor-bearing mice induced TIL entrance into S phase preceding apoptosis, showing that TIL have functional TCR-mediated signal transduction in situ. Our data demonstrate that TIL, not tumor, express both Fas and FasL, are arrested in G(1), do not secrete cytokine in situ, and, upon activation in vitro and in vivo, rapidly die by activation-induced cell death.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Cycle/immunology
- Cell Movement/immunology
- Cell Separation
- Cells, Cultured
- Immune Sera/administration & dosage
- Immunophenotyping
- In Situ Nick-End Labeling
- Injections, Intraperitoneal
- Injections, Intravenous
- Interleukin-2/administration & dosage
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Receptors, Antigen, T-Cell/immunology
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/immunology
- Tumor Cells, Cultured
- fas Receptor/physiology
Collapse
Affiliation(s)
- S Radoja
- Department of Cell Biology and Kaplan Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
34
|
Seeley BM, Barthel SW, To WC, Kjaergaard J, Shu S, Plautz GE. Potent effector function of tumor-sensitized L-selectin(low) T cells against subcutaneous tumors requires LFA-1 co-stimulation. Otolaryngol Head Neck Surg 2001; 124:436-41. [PMID: 11283503 DOI: 10.1067/mhn.2001.114253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Animal tumor models have demonstrated that adoptive transfer of tumor-draining lymph node (TDLN) T lymphocytes can cure established tumors in many anatomic sites. However, subcutaneous tumors are relatively refractory and have required maximally tolerated doses of cells. The goals of this study were to determine whether a subset of TDLN T lymphocytes varying in expression of the cell adhesion molecule L-selectin (CD62L) had augmented therapeutic efficacy and to determine the co-stimulatory requirements for trafficking and anti-tumor effector function. STUDY DESIGN TDLNs were recovered from mice bearing progressive MCA 205 fibrosarcomas, and the T lymphocytes were segregated into CD62L(low) and CD62L(high) subsets and activated ex vivo with anti-CD3 mAb and IL-2. Mice bearing established subcutaneous MCA 205 tumors were treated with activated T cell subsets and in some experiments with additional mAb against cell adhesion molecules. RESULTS Adoptive transfer of as few as 5 x 10(6) activated cells cured mice bearing 3-day subcutaneous MCA 205 tumors initiated with 6 x 10(6) cells, and the tumors demonstrated a dense infiltrate of CD62L(low) cells. In marked contrast, adoptive transfer of 10 times as many T cells derived from the reciprocal CD62L(high) compartment had no effect on tumor growth. The effector function of the CD62L(low) T cells was clearly dependent on co-stimulation through the cell adhesion molecule LFA-1, because anti-LFA-1 mAb completely abrogated the anti-tumor reactivity of the transferred cells against subcutaneous tumors and inhibited tumor infiltration. In contrast, blockade of ICAM-1, VLA-4, or VCAM-1 had no inhibitory effect on the anti-tumor function. CONCLUSION These studies demonstrate the high therapeutic activity of the CD62L(low) subset of tumor-draining LN T cells against subcutaneous tumors, a relatively refractory site, and confirm the essential role of LFA-1 for effector T cell function. SIGNIFICANCE Identification of the phenotype and requirements for effector function of T lymphocytes sensitized to tumor antigens has implications for clinical trials of adoptive immunotherapy for head and neck carcinoma using a similar approach.
Collapse
Affiliation(s)
- B M Seeley
- Center for Surgery Research, the Cleveland Clinic Foundation, OH, USA
| | | | | | | | | | | |
Collapse
|
35
|
Smilowitz HM, Micca PL, Nawrocky MM, Slatkin DN, Tu W, Coderre JA. The combination of boron neutron-capture therapy and immunoprophylaxis for advanced intracerebral gliosarcomas in rats. J Neurooncol 2001; 46:231-40. [PMID: 10902854 DOI: 10.1023/a:1006409721365] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary human brain tumor. About 7000 new cases are diagnosed yearly in the USA and GBM is almost invariably fatal within a few years after it is diagnosed. Despite current neurosurgical and radiotherapeutic tumor cytoreduction methods, in most cases occult foci of tumor cells infiltrate surrounding brain tissues and cause recurrent disease. Therefore the combination of neurosurgical and radiotherapeutic debulking methods with therapies to inhibit occult GBM cells should improve prognosis. In this study we have combined boron neutron-capture therapy (BNCT), a novel binary radiotherapeutic treatment modality that selectively irradiates tumor tissue and largely spares normal brain tissue, with immunoprophylaxis, a form of active immunization initiated soon after BNCT treatment, to treat advanced, clinically relevantly-sized brain tumors in rats. Using a malignant rat glioma model of high immunogenicity, the 9L gliosarcoma, we have shown that about half of the rats that would have died after receiving BNCT debulking alone, survived after receiving BNCT plus immunoprophylaxis. Further, most of the surviving rats display immunological-based resistance to recurrent 9LGS growth six months or more after treatment. To our knowledge this study represents the first time BNCT and immunoprophylaxis have been combined to treat advanced brain tumors in rats.
Collapse
Affiliation(s)
- H M Smilowitz
- Department of Pharmacology, University of Connecticut Health Center, Farmington 06030-6125, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Peng L, Krauss JC, Plautz GE, Mukai S, Shu S, Cohen PA. T cell-mediated tumor rejection displays diverse dependence upon perforin and IFN-gamma mechanisms that cannot be predicted from in vitro T cell characteristics. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:7116-24. [PMID: 11120842 DOI: 10.4049/jimmunol.165.12.7116] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Experimental pulmonary metastases have been successfully treated by adoptive transfer of tumor-sensitized T cells from perforin knockout (KO) or Fas/APO-1 ligand(KO) mice, suggesting a prominent role for secretion of cytokines such as IFN-gamma. In the present study we confirmed that rejection of established methylcholanthrene-205 (MCA-205) pulmonary metastases displayed a requirement for T cell IFN-gamma expression. However, this requirement could be obviated by transferring larger numbers of tumor-sensitized IFN-gamma (KO) T cells or by immunosensitizing sublethal irradiation (500 rad) of the host before adoptive therapy. Extrapulmonary tumors (MCA-205 s.c. and intracranial) that required adjunct sublethal irradiation for treatment efficacy also displayed no requirement for host or T cell expression of IFN-gamma. Nonetheless, rejection of MCA-205 s.c. tumors and i.p. EL-4 tumors, but not MCA-205 pulmonary or intracranial tumors, displayed a significant requirement for T cell perforin expression (i.e., CTL participation). The capacity of T cells to lyse tumor targets and secrete IFN-gamma in vitro before adoptive transfer was nonpredictive of the roles of these activities in subsequent tumor rejection. Adoptive therapy studies employing KO mice are therefore indispensable for revealing a diversity of tumor rejection mechanisms that may lack in vitro correlation due to delays in their induction. Seemingly contradictory KO data from different studies are reconciled by the capacity of anti-tumor T cells to rely on alternative mechanisms when treated in larger numbers, the variable participation of CTL at different anatomic locations of tumor, and the apparent capacity of sublethal irradiation to provide a therapeutic alternative to host or T cell IFN-gamma production.
Collapse
MESH Headings
- Animals
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- Dose-Response Relationship, Immunologic
- Female
- Graft Rejection/genetics
- Graft Rejection/immunology
- Graft Rejection/radiotherapy
- Immunotherapy, Adoptive
- Injections, Intraperitoneal
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Interferon-gamma/radiation effects
- Lung Neoplasms/immunology
- Lung Neoplasms/radiotherapy
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymph Nodes/pathology
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/transplantation
- Lymphoma/genetics
- Lymphoma/immunology
- Lymphoma/radiotherapy
- Lymphoma/therapy
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Transplantation
- Perforin
- Pore Forming Cytotoxic Proteins
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/radiotherapy
- Sarcoma, Experimental/therapy
- Tumor Cells, Cultured/transplantation
- Whole-Body Irradiation
Collapse
Affiliation(s)
- L Peng
- Center for Surgery Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Sloan AE, Dansey R, Zamorano L, Barger G, Hamm C, Diaz F, Baynes R, Wood G. Adoptive immunotherapy in patients with recurrent malignant glioma: preliminary results of using autologous whole-tumor vaccine plus granulocyte-macrophage colony–stimulating factor and adoptive transfer of anti-CD3–activated lymphocytes. Neurosurg Focus 2000; 9:e9. [PMID: 16817692 DOI: 10.3171/foc.2000.9.6.10] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
This trial was designed to determine the ability of autologous whole–tumor cell vaccines to induce cell-mediated immune responses in patients with recurrent malignant glioma, as well as to determine whether combining such vaccination with adoptive transfer of in vitro activated T lymphocytes prolongs patient survival.
Methods
Nineteen patients with recurrent malignant glioma, in whom previous external beam radiotherapy and at least one course of chemotherapy had failed were vaccinated twice with irradiated autologous whole tumor cells by using granulocyte-marcrophage colony–stimulating factor as an adjuvant. Patients then underwent leukapheresis followed by adoptive transfer of peripheral blood lymphocytes activated in vitro with anti-CD3 and interleukin-2. In vivo immune response, radiological response, clinical outcome, and survival were monitored.
Seventeen patients developed a delayed-type hypersensitivity (DTH) response to vaccination that appeared to be directed against the autologous tumor. In eight patients there was radiological evidence of a response and in five there was evidence of clinical improvement. Median survival was 12 months (range 6–28 months), and both the presence of a DTH response and the radiological response correlated with survival (p < 0.02 and p < 0.04, respectively).
Conclusions
These preliminary results suggest that autologous whole–tumor cell vaccines induce a cell-mediated immune response, which appears to be tumor specific in most patients. Furthermore, vaccination combined with adoptive immunotherapy with in vitro activated cells may induce a radiologically demonstrated tumor response and improved survival despite a condition of advanced disease and immunosuppression resulting from previous treatment or tumor burden. Further studies of immunotherapy are warranted.
Collapse
Affiliation(s)
- A E Sloan
- Department of Neurosurgery, Karmanos Cancer Institute, Detroit, Michigan, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Plautz GE, Mukai S, Cohen PA, Shu S. Cross-presentation of tumor antigens to effector T cells is sufficient to mediate effective immunotherapy of established intracranial tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:3656-62. [PMID: 11034369 DOI: 10.4049/jimmunol.165.7.3656] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The systemic adoptive transfer of tumor-sensitized T cells, activated ex vivo, can eliminate established intracranial tumors. Regression of MHC class II negative MCA 205 fibrosarcomas occurs optimally following adoptive transfer of both CD4 and CD8 tumor-sensitized T cells, indicating an important function for tumor-infiltrating APC. Here, we demonstrate that during an effector response, indirect presentation of tumor Ags to transferred T cells is sufficient to mediate intracranial tumor regression. BALB/c --> CB6F1 (H-2bxd) bone marrow chimeras were challenged with the MCA 205 fibrosarcoma (H-2b). The tumor grew progressively in the H-2b-tolerant chimeras and stimulated an immune response in tumor-draining lymph nodes. Tumor-sensitized lymph node T cells were activated ex vivo with anti-CD3 and IL-2, then adoptively transferred to sublethally irradiated BALB/c or C57BL/6 recipients bearing established intracranial MCA 205 tumors. The transferred T cells eradicated MCA 205 tumors in BALB/c recipients and demonstrated tumor specificity, but had no therapeutic efficacy in the C57BL/6 recipients. These data establish that tumor-associated host cell constituents provide sufficient Ag presentation to drive effector T cell function in the complete absence of direct tumor recognition. This effector mechanism has an evident capacity to remain operative in circumstances of immune escape, where the tumor does not express the relevant MHC molecules, and may have importance even at times when direct CTL recognition also remains operative.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen Presentation/genetics
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Brain Neoplasms/immunology
- Brain Neoplasms/pathology
- Brain Neoplasms/therapy
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- Fibrosarcoma/immunology
- Fibrosarcoma/pathology
- Fibrosarcoma/therapy
- Histocompatibility Antigens Class II/biosynthesis
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Regression, Spontaneous
- Radiation Chimera/immunology
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/pathology
- Sarcoma, Experimental/therapy
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- G E Plautz
- Center for Surgery Research, The Cleveland Clinic Foundation, OH 44195, USA.
| | | | | | | |
Collapse
|
39
|
Wood GW, Holladay FP, Turner T, Wang YY, Chiga M. A pilot study of autologous cancer cell vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in patients with recurrent grade III/IV astrocytoma. J Neurooncol 2000; 48:113-20. [PMID: 11083074 DOI: 10.1023/a:1006456421177] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The study objectives were to determine; (1) whether activated T cells could be generated from peripheral blood of patients immunized with their own cancer cells, (2) whether adoptive transfer of the activated T cells to patients had toxic effects and (3) whether the infused cells produced clinical responses. Study patients had recurrent, surgically accessible grade III/IV astrocytomas. The patients were tapered off steroids after total surgical resection and immunized with autologous cancer cells plus Bacillus, Calmette and Guerin (BCG). Peripheral blood mononuclear cells were activated with anti-CD3, expanded with interleukin-2 (IL-2) and reinfused to patients. The number of activated T cells that was given back to patients varied between 10(10) and 10(11). Side effects that were observed following immunization and adoptive cell transfer included mainly transient flu-like symptoms. One patient's tumor partially regressed, but there was no effect on survival. Two other patients' tumors regressed, and the patients are apparently disease-free more than 5 and 4 years later. The other six patients' tumors were apparently unaffected by the treatment. Patient age, tumor grade and CD4/CD8 composition of infused cells were positively correlated with clinical responses. Cellular immunotherapy is feasible and is associated with minimal toxicity. Additional appropriately controlled studies will be required to determine whether cellular immunotherapy could be used as a treatment for central nervous system malignancy. Additional studies also will be required to determine the underlying immunological mechanisms.
Collapse
Affiliation(s)
- G W Wood
- Department of Pathology, University of Kansas Medical Center, Kansas City, USA.
| | | | | | | | | |
Collapse
|
40
|
Plautz GE, Bukowski RM, Novick AC, Klein EA, Kursh ED, Olencki TE, Yetman RJ, Pienkny A, Sandstrom K, Shu S. T-cell adoptive immunotherapy of metastatic renal cell carcinoma. Urology 1999; 54:617-23; discussion 623-4. [PMID: 10510917 DOI: 10.1016/s0090-4295(99)00303-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To determine the feasibility and toxicity of the adoptive transfer of ex vivo-activated T lymphocytes that have been sensitized to autologous tumor vaccine in vivo. METHODS Twenty patients with extensive metastatic renal cell carcinoma received systemic adoptive immunotherapy with autologous T cells in the absence of conjunctional interleukin-2 (IL-2) administration. Patients were vaccinated intradermally with irradiated autologous tumor cells and granulocyte-macrophage colony-stimulating factor as an adjuvant to stimulate an immune response. Inguinal lymph nodes draining the vaccine site were surgically removed, and the cells were stimulated with staphylococcal enterotoxin A followed by expansion in 60 IU/mL IL-2, and in some cases additionally stimulated with anti-CD3 monoclonal antibody and IL-2, to obtain a large number of cells. RESULTS The staphylococcal enterotoxin A/IL-2 activation induced vigorous proliferation of T cells (median expansion 26-fold) that were a mixture of CD4 and CD8 T lymphocytes. Activated cells were infused intravenously at doses ranging from 2x10(9) to 9.5x10(10). There was minimal toxicity consisting of grade 1 or 2 fever and nausea, and the entire treatment was delivered as outpatient therapy. One patient had a partial response, one had a mixed response, and 8 had stable disease lasting at least 5 months. CONCLUSIONS Adoptive transfer of ex vivo-activated, tumor vaccine-primed lymph node cells is feasible and is associated with minimal toxicity when used alone. These results warrant further study in a Phase II trial.
Collapse
Affiliation(s)
- G E Plautz
- Center for Surgery Research, Experimental Therapeutics Program and Department of Urology, Cleveland Clinic Foundation, Ohio, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mukai S, Kagamu H, Shu S, Plautz GE. Critical role of CD11a (LFA-1) in therapeutic efficacy of systemically transferred antitumor effector T cells. Cell Immunol 1999; 192:122-32. [PMID: 10087180 DOI: 10.1006/cimm.1998.1439] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The systemic adoptive transfer of activated T cells, derived from tumor-draining lymph nodes (LNs), mediates the regression of established tumors. In this study, the requirement of cell adhesion molecules, CD11a/CD18 (LFA-1), CD54 (ICAM-1), CD49d/CD29 (VLA-4), and CD106 (VCAM-1), for T cell infiltration into tumors and antitumor function was investigated. Administration of anti-CD11a mAb completely abrogated the efficacy of adoptive immunotherapy for both intracranial and pulmonary metastatic MCA 205 fibrosarcomas. In contrast, adoptive immunotherapy was effective in animals treated with anti-CD49d mAb, anti-CD106 mAb, anti-CD54 mAb, or in CD54 knockout recipients. Trafficking of transferred cells to the intracranial tumor was not affected by any of the mAb. However, the tumor-specific secretion of IFN-gamma by activated LN T cells was suppressed by anti-CD11a mAb or anti-CD54 mAb. To account for the different effects of CD11a and CD54 blockade in vivo, an additional CD11a/CD18 ligand, CD102 (ICAM-2), was demonstrated on tumor-associated macrophages but not on tumor cells. These results show that CD11a mediates a critical function in interactions between effector T cells, tumor cells, and host accessory cells in situ leading to tumor regression.
Collapse
Affiliation(s)
- S Mukai
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
42
|
Plautz GE, Barnett GH, Miller DW, Cohen BH, Prayson RA, Krauss JC, Luciano M, Kangisser DB, Shu S. Systemic T cell adoptive immunotherapy of malignant gliomas. J Neurosurg 1998; 89:42-51. [PMID: 9647171 DOI: 10.3171/jns.1998.89.1.0042] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT To determine the feasibility, toxicity, and potential therapeutic benefits of systemic adoptive immunotherapy, 10 patients with progressive primary or recurrent malignant glioma received this treatment. Adoptive immunotherapy, the transfer of immune T lymphocytes, is capable of mediating the regression of experimental brain tumors in animal models. In animal models, lymph nodes (LNs) that drain the tumor vaccine site are a rich source of tumor-immune T cells. METHODS In this clinical study, patients were inoculated intradermally with irradiated autologous tumor cells and granulocyte macrophage-colony stimulating factor as an adjuvant. Cells from draining inguinal LNs, surgically resected 7 days after vaccination, were stimulated sequentially with staphylococcal enterotoxin A and anti-CD3, and a low dose of interleukin-2 (60 IU/ml) was used to expand the stimulated cells. The maximum cell proliferation was 350-fold over 10 days of culture. The activated cells were virtually all T cells consisting of various proportions of CD4 and CD8 cells. These cells were given to patients by intravenous infusion at doses ranging from 9 x 10(8) to 1.5 x 10(11). There were no Grade 3 or 4 toxicities associated with the treatment. Following T-cell transfer therapy, radiographic regression that lasted at least 6 months was demonstrated in two patients with recurrent tumors. One patient demonstrated stable disease that has lasted for more than 17 months. The remaining patients had progressive disease; however, four of the eight patients with recurrent tumor remain alive more than 1 year after surgery for recurrence. Three patients required intervention with corticosteroid agents or additional surgery approximately 1 month following cell transfer. CONCLUSIONS These intriguing clinical observations warrant further trials to determine whether this approach can provide therapeutic benefits and improve survival.
Collapse
Affiliation(s)
- G E Plautz
- Department of Neurological Surgery, Center for Surgery Research, The Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Plautz GE, Barnett GH, Miller DW, Cohen BH, Prayson RA, Krauss JC, Luciano M, Shu S. Activated lymph node T cells for systemic adoptive immunotherapy of malignant glioma. Neurosurg Focus 1997. [DOI: 10.3171/foc.1997.3.5.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ten patients with progressive primary or recurrent malignant glioma received systemic adoptive immunotherapy to determine the feasibility, toxicity, and potential therapeutic benefits of this treatment. Adoptive immunotherapy, the transfer of immune T lymphocytes, is capable of mediating the regression of experimental brain tumors in animal models. A rich source of tumor-immune T cells are lymph nodes (LNs) draining the tumor vaccine site. In this clinical study, patients were inoculated intradermally with irradiated autologous tumor cells and granulocyte-macrophage colony stimulating factor as an adjuvant. Cells from draining inguinal LNs, surgically resected 7 days after vaccination, were stimulated sequentially with staphylococcal enterotoxin A and anti-CD3, and a low dose of interleukin-2 (60 IU/ml) was used to expand the stimulated cells. The maximum cell proliferation was 350-fold over 10 days of culture. The activated cells were virtually all T cells consisting of various proportions of CD4 and CD8 cells. These cells were given to patients by intravenous infusion at doses ranging from 9 X 108 to 1.5 X 1011. There were no Grade 3 or 4 toxicities associated with the treatment. Following T cell transfer therapy, radiographic regression that lasted at least 4 months was demonstrated in three patients with recurrent tumors, and four patients remain alive more than 11 months after surgery. The remaining patients had progressive disease, and three patients required intervention with corticosteroid agents or additional surgery approximately 1 month following cell transfer. These findings demonstrate that adoptive immunotherapy can be administered in patients with glioma without causing significant toxicity. It appears that this experimental regimen can provide therapeutic benefits for some patients.
Collapse
|