1
|
Cui M, Zhou M, Zhou L, Zhou G, Liu Y. Tertiary lymphoid structures achieve 'cold' to 'hot' transition by remodeling the cold tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189312. [PMID: 40189114 DOI: 10.1016/j.bbcan.2025.189312] [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: 06/11/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Immune checkpoint blockade (ICB) therapies have demonstrated significant clinical efficacy in immune-infiltrated tumors such as melanoma and non-small cell lung cancer. However, "cold tumors"-including ovarian cancer, pancreatic cancer, and gliomas-exhibit insufficient immune infiltration, leading to poor therapeutic responses to ICBs and limited improvement in patient prognosis. Recent studies have shown that tumor-associated tertiary lymphoid structures (TLSs) can induce strong local immune responses within the tumor microenvironment (TME), serving as important biological markers for predicting ICB therapy efficacy. Notably, preclinical and clinical studies on cold tumors have confirmed that TLSs can potently enhance ICB efficacy through TME remodeling-a breakthrough that has attracted considerable attention. Here, we systematically examine the immunological profile of cold tumors and decipher the mechanistic basis for their impaired immune cell infiltration. We further delineate the distinctive features of tumor-associated TLSs in generating antitumor immunity and establish criteria for their identification. Significantly, we emphasize the unique capability of TLSs to reprogram the immunosuppressive tumor microenvironment characteristic of cold tumors. Based on these insights, we evaluate clinical evidence supporting TLS-mediated enhancement of ICB efficacy and discuss emerging strategies for exogenous TLSs induction.
Collapse
Affiliation(s)
- Mengke Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Mengfan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Lu Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiangya Road, Changsha, Hunan 410008, PR China.
| | - Yingzi Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China.
| |
Collapse
|
2
|
Stasiak K, Stevens AD, Bolte AC, Curley CT, Perusina Lanfranca M, Lindsay RS, Eyo UB, Lukens JR, Price RJ, Bullock TNJ, Engelhard VH. Differential T cell accumulation within intracranial and subcutaneous melanomas is associated with differences in intratumoral myeloid cells. Cancer Immunol Immunother 2024; 74:10. [PMID: 39487854 PMCID: PMC11531463 DOI: 10.1007/s00262-024-03832-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 11/04/2024]
Abstract
Patients with metastatic brain melanomas (MBM) experience shorter-lasting survival than patients with extracranial metastases, and this is associated with a higher fraction of dysfunctional CD8 T cells. The goal of this study was to understand the underlying cause of T cell dysfunction in MBM. To accomplish this, we compared murine B16 melanomas implanted intracranially (IC) or subcutaneously (SC). CD8 T cell activation was not altered, but representation in IC tumors was lower. Transferred activated or naïve CD8 T cells accumulated in similar numbers in both tumors, suggesting that the vasculature does not differentially impair T cell presence. Surprisingly, we found no evidence for T cell activation in draining lymph nodes of SC or IC tumor-bearing mice, consistent with the fact that dendritic cells (DC) that had acquired tumor antigen showed an immature phenotype. Instead, T cell activation occurred within both tumors, where the majority of tumor antigen+ myeloid cells were found. While, the numbers of intratumoral DC were comparable, those in IC tumors acquired less tumor antigen, and were alternatively matured based on upregulation of MHCII without upregulation of CD86. Additionally, in IC tumors, the largest population of tumor antigen+ myeloid cells were microglia. However, their presence did not influence either antigen acquisition or the phenotype of other myeloid cell populations. Overall, our data suggest that diminished representation of CD8 T cells in IC tumors is a consequence of alternatively matured DC and/or microglia that induce distinctly activated T cells, which ultimately fail to continue to accumulate inside the tumor.
Collapse
Affiliation(s)
- Katarzyna Stasiak
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Aaron D Stevens
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Ashley C Bolte
- Department of Neuroscience, University of Virginia, Charlottesville, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, USA
| | - Colleen T Curley
- Department of Biomedical Engineering, University of Virgnia, Charlottesville, VA, USA
| | - Mirna Perusina Lanfranca
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Robin S Lindsay
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Ukpong B Eyo
- Department of Neuroscience, University of Virginia, Charlottesville, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, USA
| | - John R Lukens
- Department of Neuroscience, University of Virginia, Charlottesville, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virgnia, Charlottesville, VA, USA
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, USA
| | | | - Victor H Engelhard
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA.
| |
Collapse
|
3
|
Zhang Q, Zhang J, Tian Y, Wang J, Jin G, Liu F. Ki67-targeted oncolytic adenovirus expressing IL-15 improves intratumoral T cell infiltration and PD-L1 expression in glioblastoma. Virology 2023; 587:109885. [PMID: 37738842 DOI: 10.1016/j.virol.2023.109885] [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: 05/05/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023]
Abstract
Glioblastoma (GBM) is a devastating malignant brain tumor. Current therapeutic strategies targeting tumor cells have limited efficacy owing to the immunosuppressive microenvironment. Previous work demonstrated that the targeted Ad5-Ki67/IL-15 could specifically kill tumor cells and decrease the angiogenic capacity in vitro. However, the efficacy of this virus in vivo and its effect on the tumor microenvironment (TME) has not been elucidated. In this study, we found that the Ad5-Ki67/IL-15 treatment down-regulated PD-L1 expression of glioma cells. More importantly, Ad5-Ki67/IL-15 also remodeled the tumor microenvironment via increasing intratumoral T cell infiltration and PD-L1 improvement in a GBM model, as well as the increase of antitumor cytokines, thereby improving the efficacy of GBM treatment. Furthermore, a combination of Ad5-Ki67/IL-15 with PD-L1 blockade significantly inhibits tumor growth in the GBM model. These results provide new insight into the therapeutic effects of targeted oncolytic Ad5-Ki67/IL-15 in patients with GBM, indicating potential clinical applications.
Collapse
Affiliation(s)
- Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China; Beijing Laboratory of Biomedical Materials, Beijing, 100070, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China; Beijing Laboratory of Biomedical Materials, Beijing, 100070, China
| | - Yifu Tian
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China; Beijing Laboratory of Biomedical Materials, Beijing, 100070, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China; Beijing Laboratory of Biomedical Materials, Beijing, 100070, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China; Beijing Laboratory of Biomedical Materials, Beijing, 100070, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China; Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China; Beijing Laboratory of Biomedical Materials, Beijing, 100070, China.
| |
Collapse
|
4
|
Liu X, Zhao Z, Dai W, Liao K, Sun Q, Chen D, Pan X, Feng L, Ding Y, Wei S. The Development of Immunotherapy for the Treatment of Recurrent Glioblastoma. Cancers (Basel) 2023; 15:4308. [PMID: 37686584 PMCID: PMC10486426 DOI: 10.3390/cancers15174308] [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: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023] Open
Abstract
Recurrent glioblastoma (rGBM) is a highly aggressive form of brain cancer that poses a significant challenge for treatment in neuro-oncology, and the survival status of patients after relapse usually means rapid deterioration, thus becoming the leading cause of death among patients. In recent years, immunotherapy has emerged as a promising strategy for the treatment of recurrent glioblastoma by stimulating the body's immune system to recognize and attack cancer cells, which could be used in combination with other treatments such as surgery, radiation, and chemotherapy to improve outcomes for patients with recurrent glioblastoma. This therapy combines several key methods such as the use of monoclonal antibodies, chimeric antigen receptor T cell (CAR-T) therapy, checkpoint inhibitors, oncolytic viral therapy cancer vaccines, and combination strategies. In this review, we mainly document the latest immunotherapies for the treatment of glioblastoma and especially focus on rGBM.
Collapse
Affiliation(s)
- Xudong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Zihui Zhao
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering Research, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China;
| | - Qi Sun
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Dongjiang Chen
- Division of Neuro-Oncology, USC Keck Brain Tumor Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Lishuang Feng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Ying Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Shiyou Wei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
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
|
6
|
Rong L, Li N, Zhang Z. Emerging therapies for glioblastoma: current state and future directions. J Exp Clin Cancer Res 2022; 41:142. [PMID: 35428347 PMCID: PMC9013078 DOI: 10.1186/s13046-022-02349-7] [Citation(s) in RCA: 229] [Impact Index Per Article: 76.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/26/2022] [Indexed: 04/15/2023] Open
Abstract
Glioblastoma (GBM) is the most common high-grade primary malignant brain tumor with an extremely poor prognosis. Given the poor survival with currently approved treatments for GBM, new therapeutic strategies are urgently needed. Advances in decades of investment in basic science of glioblastoma are rapidly translated into innovative clinical trials, utilizing improved genetic and epigenetic profiling of glioblastoma as well as the brain microenvironment and immune system interactions. Following these encouraging findings, immunotherapy including immune checkpoint blockade, chimeric antigen receptor T (CAR T) cell therapy, oncolytic virotherapy, and vaccine therapy have offered new hope for improving GBM outcomes; ongoing studies are using combinatorial therapies with the aim of minimizing adverse side-effects and augmenting antitumor immune responses. In addition, techniques to overcome the blood-brain barrier (BBB) for targeted delivery are being tested in clinical trials in patients with recurrent GBM. Here, we set forth the rationales for these promising therapies in treating GBM, review the potential novel agents, the current status of preclinical and clinical trials, and discuss the challenges and future perspectives in glioblastoma immuno-oncology.
Collapse
Affiliation(s)
- Liang Rong
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ni Li
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Zhang
- Key Laboratory of Brain, Cognition and Education Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China.
| |
Collapse
|
7
|
Tang B, Guo ZS, Bartlett DL, Yan DZ, Schane CP, Thomas DL, Liu J, McFadden G, Shisler JL, Roy EJ. Synergistic Combination of Oncolytic Virotherapy and Immunotherapy for Glioma. Clin Cancer Res 2020; 26:2216-2230. [PMID: 32019860 DOI: 10.1158/1078-0432.ccr-18-3626] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2019] [Accepted: 01/30/2020] [Indexed: 01/10/2023]
Abstract
PURPOSE We hypothesized that the combination of a local stimulus for activating tumor-specific T cells and an anti-immunosuppressant would improve treatment of gliomas. Virally encoded IL15Rα-IL15 as the T-cell activating stimulus and a prostaglandin synthesis inhibitor as the anti-immunosuppressant were combined with adoptive transfer of tumor-specific T cells. EXPERIMENTAL DESIGN Two oncolytic poxviruses, vvDD vaccinia virus and myxoma virus, were each engineered to express the fusion protein IL15Rα-IL15 and a fluorescent protein. Viral gene expression (YFP or tdTomato Red) was confirmed in the murine glioma GL261 in vitro and in vivo. GL261 tumors in immunocompetent C57BL/6J mice were treated with vvDD-IL15Rα-YFP vaccinia virus or vMyx-IL15Rα-tdTr combined with other treatments, including vaccination with GARC-1 peptide (a neoantigen for GL261), rapamycin, celecoxib, and adoptive T-cell therapy. RESULTS vvDD-IL15Rα-YFP and vMyx-IL15Rα-tdTr each infected and killed GL261 cells in vitro. In vivo, NK cells and CD8+ T cells were increased in the tumor due to the expression of IL15Rα-IL15. Each component of a combination treatment contributed to prolonging survival: an oncolytic virus, the IL15Rα-IL15 expressed by the virus, a source of T cells (whether by prevaccination or adoptive transfer), and prostaglandin inhibition all synergized to produce elimination of gliomas in a majority of mice. vvDD-IL15Rα-YFP occasionally caused ventriculitis-meningitis, but vMyx-IL15Rα-tdTr was safe and effective, causing a strong infiltration of tumor-specific T cells and eliminating gliomas in 83% of treated mice. CONCLUSIONS IL15Rα-IL15-armed oncolytic poxviruses provide potent antitumor effects against brain tumors when combined with adoptive T-cell therapy, rapamycin, and celecoxib.
Collapse
Affiliation(s)
- Bingtao Tang
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Zong Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David Z Yan
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Claire P Schane
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Diana L Thomas
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Grant McFadden
- Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Joanna L Shisler
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Edward J Roy
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
8
|
Ladányi A, Tímár J. Immunologic and immunogenomic aspects of tumor progression. Semin Cancer Biol 2019; 60:249-261. [PMID: 31419526 DOI: 10.1016/j.semcancer.2019.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
Tumor progression to metastatic disease is characterized by continuous genetic alterations due to instability of the genome. Immune sensitivity was found to be linked to tumor mutational burden (TMB) and the resulting amount of neoantigens. However, APOBEC activity resulting in increase in TMB causes immune evasion. On the other hand, clonal or acquired genetic loss of HLA class I also hampers immune sensitivity of tumors. Rare amplification of the PD-L1 gene in cancers may render them sensitive to immune checkpoint inhibitors but involvement of broader regions of chromosome 9p may ultimately lead again to immune evasion due to inactivation of the IFN-γ signaling pathway. Such genetic changes may occur not only in the primary tumor but at any phase of progression: in lymphatic as well as in visceral metastases. Accordingly, it is rational to monitor these changes continuously during disease progression similar to target therapies. Moreover, beside temporal variability, genomic features of tumors such as mutation profiles, as well as the tumor immune microenvironment also show considerable inter- and intratumoral spatial heterogeneity, suggesting the necessity of multiple sampling in biomarker studies.
Collapse
Affiliation(s)
| | - József Tímár
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
9
|
Zhong M, Zhong C, Hu P, Cui W, Wang G, Gao H, Liu C, Liu Z, Li Z, Li C, Gohda E. Restoration of stemness-high tumor cell-mediated suppression of murine dendritic cell activity and inhibition of tumor growth by low molecular weight oyster polysaccharide. Int Immunopharmacol 2018; 65:221-232. [PMID: 30321818 DOI: 10.1016/j.intimp.2018.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/24/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play key regulatory roles in tumor immunity: increased activity of DCs infiltrating tumor tissues leads to enhancement of tumor immunity. Functions of DCs are also modulated by tumor cell-derived factors. Here, we investigated the effects of low molecular weight oyster polysaccharide (LMW-OPS) on differentiation and function of bone marrow-derived DCs (BMDCs) exposed to a conditioned medium (CM) obtained from spheres of stemness-high colorectal cancer cell lines CMT93 and CT26. The CM containing a detectable level of TGF-β1 was found to down-regulate the surface expression of major histocompatibility complex class II of BMDCs and to inhibit the potency of BMDCs to stimulate T cells. Those suppressions were partly restored and completely restored by addition of anti-TGF-β1 and LMW-OPS, respectively. Production of IFN-γ during allogeneic T cell responses was inhibited by the CM, whereas production of TGF-β1 was augmented by the CM. The IFN-γ profile was also reversed by addition of LMW-OPS. Nuclear translocation of β-catenin, but not that of NF-κB p65, was induced by TGF-β1. NF-κB p65 nuclear translocation, but not β-catenin nuclear translocation, was induced by LMW-OPS. Intraperitoneal injection of LMW-OPS significantly suppressed tumor growth in syngeneic tumor models using CMT93 and CT26 sphere cells, whereas it had no inhibitory effect on the proliferation of either cell line. The results demonstrated that LMW-OPS relieved stemness-high tumor cell-mediated suppression of BMDC function and indicated the in vivo anti-tumor activity of LMW-OPS in which re-stimulation of the activity of DCs infiltrating tumor tissues is presumed to be involved.
Collapse
Affiliation(s)
- Ming Zhong
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China.
| | - Cheng Zhong
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Pei Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Wen Cui
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Guanghui Wang
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Huijei Gao
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Chao Liu
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Zhiqiang Liu
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Zhihua Li
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Chunxia Li
- Institute of Tumor Pharmacology, Jining Medical College, Rizhao, China
| | - Eiichi Gohda
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
10
|
Wang YG, Long J, Shao DC, Song H. Hyperbaric oxygen inhibits production of CD3+ T cells in the thymus and facilitates malignant glioma cell growth. J Int Med Res 2018; 46:2780-2791. [PMID: 29785863 PMCID: PMC6124287 DOI: 10.1177/0300060518767796] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective Hyperbaric oxygen (HBO) is an emerging complementary alternative medical approach in glioma treatment. However, its mode of action is unknown, so this was investigated in the present study. Methods We constructed an intracranial glioma model of congenic C57BL/6J mice. Glioma growth under HBO stimulation was assessed by bioluminescent imaging and magnetic resonance imaging. Flow cytometry assessed direct effects of HBO on reactive oxygen species (ROS) signaling of transplanted glioma cells and organs, and quantified mature T cells and subgroups in tumors, the brain, and blood. Results HBO promoted the growth of transplanted GL261-Luc glioma in the intracranial glioma mouse model. ROS signaling of glioma cells and brain cells was significantly downregulated under HBO stimulation, but thymus ROS levels were significantly upregulated. CD3+ T cells were significantly downregulated, while both Ti/Th cells (CD3+CD4+) and Ts/Tc cells (CD3+CD8+) were inhibited in tumors of the HBO group. The percentage of regulatory T cells in Ti/Th (CD3+CD4+) cells was elevated in the tumors and thymuses of the HBO group. Conclusion HBO induced ROS signaling in the thymus, inhibited CD3+ T cell generation, and facilitated malignant glioma cell growth in vivo in the intracranial glioma mouse model.
Collapse
Affiliation(s)
- Yong-Gang Wang
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jiang Long
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Dong-Chuan Shao
- 2 Department of Neurosurgery, First People's Hospital of Kunming, Kunming, Yunnan 650032, China
| | - Hai Song
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| |
Collapse
|
11
|
Abstract
OPINION STATEMENT Immune checkpoint inhibitors have changed the landscape of cancer immunotherapy and are being integrated into the standard of care for a variety of solid and hematologic malignancies. Glioblastoma (GBM) is the most common primary malignant brain tumor in adults and carries a grave prognosis despite advances in surgical resection, chemotherapy, and radiation therapy. Implementing immunotherapy for brain tumors mandates additional considerations due to the unique structural and immunologic milieu of the central nervous system (CNS). Nevertheless, strong data from preclinical studies have driven clinical trials of immune checkpoint blockade for newly diagnosed and recurrent GBM. The focus of this review is to discuss the ongoing clinical trials of checkpoint inhibitors in GBM and review the immunologic rationale for ongoing and future trial designs.
Collapse
|
12
|
De Feo D, Merlini A, Brambilla E, Ottoboni L, Laterza C, Menon R, Srinivasan S, Farina C, Garcia Manteiga JM, Butti E, Bacigaluppi M, Comi G, Greter M, Martino G. Neural precursor cell-secreted TGF-β2 redirects inflammatory monocyte-derived cells in CNS autoimmunity. J Clin Invest 2017; 127:3937-3953. [PMID: 28945200 DOI: 10.1172/jci92387] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/02/2017] [Indexed: 12/28/2022] Open
Abstract
In multiple sclerosis, the pathological interaction between autoreactive Th cells and mononuclear phagocytes in the CNS drives initiation and maintenance of chronic neuroinflammation. Here, we found that intrathecal transplantation of neural stem/precursor cells (NPCs) in mice with experimental autoimmune encephalomyelitis (EAE) impairs the accumulation of inflammatory monocyte-derived cells (MCs) in the CNS, leading to improved clinical outcome. Secretion of IL-23, IL-1, and TNF-α, the cytokines required for terminal differentiation of Th cells, decreased in the CNS of NPC-treated mice, consequently inhibiting the induction of GM-CSF-producing pathogenic Th cells. In vivo and in vitro transcriptome analyses showed that NPC-secreted factors inhibit MC differentiation and activation, favoring the switch toward an antiinflammatory phenotype. Tgfb2-/- NPCs transplanted into EAE mice were ineffective in impairing MC accumulation within the CNS and failed to drive clinical improvement. Moreover, intrathecal delivery of TGF-β2 during the effector phase of EAE ameliorated disease severity. Taken together, these observations identify TGF-β2 as the crucial mediator of NPC immunomodulation. This study provides evidence that intrathecally transplanted NPCs interfere with the CNS-restricted inflammation of EAE by reprogramming infiltrating MCs into antiinflammatory myeloid cells via secretion of TGF-β2.
Collapse
Affiliation(s)
| | | | | | | | | | - Ramesh Menon
- Immunobiology of Neurological Disorders Lab, Institute of Experimental Neurology, Division of Neuroscience, and
| | - Sundararajan Srinivasan
- Immunobiology of Neurological Disorders Lab, Institute of Experimental Neurology, Division of Neuroscience, and
| | - Cinthia Farina
- Immunobiology of Neurological Disorders Lab, Institute of Experimental Neurology, Division of Neuroscience, and
| | - Jose Manuel Garcia Manteiga
- Center for Translational Genomics and BioInformatics, San Raffaele Scientific Institute and Vita Salute San Raffaele University, Milan, Italy
| | | | | | | | - Melanie Greter
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
13
|
Parsa R, Lund H, Tosevski I, Zhang XM, Malipiero U, Beckervordersandforth J, Merkler D, Prinz M, Gyllenberg A, James T, Warnecke A, Hillert J, Alfredsson L, Kockum I, Olsson T, Fontana A, Suter T, Harris RA. TGFβ regulates persistent neuroinflammation by controlling Th1 polarization and ROS production via monocyte-derived dendritic cells. Glia 2016; 64:1925-37. [PMID: 27479807 PMCID: PMC5053226 DOI: 10.1002/glia.23033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
Intracerebral levels of Transforming Growth Factor beta (TGFβ) rise rapidly during the onset of experimental autoimmune encephalomyelitis (EAE), a mouse model of Multiple Sclerosis (MS). We addressed the role of TGFβ responsiveness in EAE by targeting the TGFβ receptor in myeloid cells, determining that Tgfbr2 was specifically targeted in monocyte‐derived dendritic cells (moDCs) but not in CNS resident microglia by using bone‐marrow chimeric mice. TGFβ responsiveness in moDCs was necessary for the remission phase since LysMCreTgfbr2fl/fl mice developed a chronic form of EAE characterized by severe demyelination and extensive infiltration of activated moDCs in the CNS. Tgfbr2 deficiency resulted in increased moDC IL‐12 secretion that skewed T cells to produce IFN‐γ, which in turn enhanced the production of moDC‐derived reactive oxygen species that promote oxidative damage and demyelination. We identified SNPs in the human NOX2 (CYBB) gene that associated with the severity of MS, and significantly increased CYBB expression was recorded in PBMCs from both MS patients and from MS severity risk allele rs72619425‐A carrying individuals. We thus identify a novel myeloid cell‐T cell activation loop active in the CNS during chronic disease that could be therapeutically targeted. GLIA 2016;64:1925–1937
Collapse
Affiliation(s)
- Roham Parsa
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Harald Lund
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Ivana Tosevski
- Clinical Immunology, University Hospital of Zurich, Switzerland
| | - Xing-Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | | | - Jan Beckervordersandforth
- Institute of Neuropathology and BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.,Department of Neuropathology, Georg-August-University Goettingen, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, and; Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Marco Prinz
- Department of Neuropathology, Georg-August-University Goettingen, Germany
| | - Alexandra Gyllenberg
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Tojo James
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Andreas Warnecke
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Jan Hillert
- Neurogenetics Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Lars Alfredsson
- Cardiovascular Epidemiology Unit, Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Adriano Fontana
- Clinical Immunology, University Hospital of Zurich, Switzerland.,Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Tobias Suter
- Clinical Immunology, University Hospital of Zurich, Switzerland.,Department of Neurology and Clinical Research Priority Program Multiple Sclerosis, University Hospital Zurich, Switzerland
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
14
|
Domingues P, González-Tablas M, Otero Á, Pascual D, Miranda D, Ruiz L, Sousa P, Ciudad J, Gonçalves JM, Lopes MC, Orfao A, Tabernero MD. Tumor infiltrating immune cells in gliomas and meningiomas. Brain Behav Immun 2016. [PMID: 26216710 DOI: 10.1016/j.bbi.2015.07.019] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tumor-infiltrating immune cells are part of a complex microenvironment that promotes and/or regulates tumor development and growth. Depending on the type of cells and their functional interactions, immune cells may play a key role in suppressing the tumor or in providing support for tumor growth, with relevant effects on patient behavior. In recent years, important advances have been achieved in the characterization of immune cell infiltrates in central nervous system (CNS) tumors, but their role in tumorigenesis and patient behavior still remain poorly understood. Overall, these studies have shown significant but variable levels of infiltration of CNS tumors by macrophage/microglial cells (TAM) and to a less extent also lymphocytes (particularly T-cells and NK cells, and less frequently also B-cells). Of note, TAM infiltrate gliomas at moderate numbers where they frequently show an immune suppressive phenotype and functional behavior; in contrast, infiltration by TAM may be very pronounced in meningiomas, particularly in cases that carry isolated monosomy 22, where the immune infiltrates also contain greater numbers of cytotoxic T and NK-cells associated with an enhanced anti-tumoral immune response. In line with this, the presence of regulatory T cells, is usually limited to a small fraction of all meningiomas, while frequently found in gliomas. Despite these differences between gliomas and meningiomas, both tumors show heterogeneous levels of infiltration by immune cells with variable functionality. In this review we summarize current knowledge about tumor-infiltrating immune cells in the two most common types of CNS tumors-gliomas and meningiomas-, as well as the role that such immune cells may play in the tumor microenvironment in controlling and/or promoting tumor development, growth and control.
Collapse
Affiliation(s)
- Patrícia Domingues
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María González-Tablas
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Álvaro Otero
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Daniel Pascual
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - David Miranda
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Laura Ruiz
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Pablo Sousa
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Juana Ciudad
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | | | - María Celeste Lopes
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Alberto Orfao
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María Dolores Tabernero
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain; Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain; Instituto de Estudios de Ciencias de la salud de Castilla y León (IECSCYL-IBSAL) and Research Unit of the University Hospital of Salamanca, Salamanca, Spain.
| |
Collapse
|
15
|
Clark AJ, Fakurnejad S, Ma Q, Hashizume R. Bioluminescence Imaging of an Immunocompetent Animal Model for Glioblastoma. J Vis Exp 2016:e53287. [PMID: 26863490 PMCID: PMC4781579 DOI: 10.3791/53287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In contrast to commonly reported human glioma xenograft animal models, GL261 murine glioma xenografts recapitulate nearly all relevant clinical and histopathologic features of the human disease. When GL261 cells are implanted intracranially in syngeneic C57BL/6 mice, the model has the added advantage of maintaining an intact immune microenvironment. Stable expression of luciferase in GL261 cells allows non-invasive cost effective bioluminescence monitoring of intracranial tumor growth. We have recently demonstrated that luciferase expression in GL261 cells does not affect the tumor growth properties, tumor cell immunomodulatory cytokine expression, infiltration of immune cells into the tumor, or overall survival of animals bearing the intracranial tumor. Therefore, it appears that the GL261 luciferase glioma model can be useful in the study of novel chemotherapeutic and immunotherapeutic modalities. Here we report the technique for generating stable luciferase expression in GL261 cells and how to study the in vitro and in vivo growth of the tumor cells by bioluminescence imaging.
Collapse
Affiliation(s)
- Aaron J Clark
- Department of Neurological Surgery, University of California San Francisco
| | - Shayan Fakurnejad
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine
| | - Quanhong Ma
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine
| | - Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine; Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine;
| |
Collapse
|
16
|
Jackson CM, Kochel CM, Nirschl CJ, Durham NM, Ruzevick J, Alme A, Francica BJ, Elias J, Daniels A, Dubensky TW, Lauer P, Brockstedt DG, Baxi EG, Calabresi PA, Taube JM, Pardo CA, Brem H, Pardoll DM, Lim M, Drake CG. Systemic Tolerance Mediated by Melanoma Brain Tumors Is Reversible by Radiotherapy and Vaccination. Clin Cancer Res 2015; 22:1161-72. [PMID: 26490306 DOI: 10.1158/1078-0432.ccr-15-1516] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 09/26/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE Immune responses to antigens originating in the central nervous system (CNS) are generally attenuated, as collateral damage can have devastating consequences. The significance of this finding for the efficacy of tumor-targeted immunotherapies is largely unknown. EXPERIMENTAL DESIGN The B16 murine melanoma model was used to compare cytotoxic responses against established tumors in the CNS and in the periphery. Cytokine analysis of tissues from brain tumor-bearing mice detected elevated TGFβ secretion from microglia and in the serum and TGFβ signaling blockade reversed tolerance of tumor antigen-directed CD8 T cells. In addition, a treatment regimen using focal radiation therapy and recombinant Listeria monocytogenes was evaluated for immunologic activity and efficacy in this model. RESULTS CNS melanomas were more tolerogenic than equivalently progressed tumors outside the CNS as antigen-specific CD8 T cells were deleted and exhibited impaired cytotoxicity. Tumor-bearing mice had elevated serum levels of TGFβ; however, blocking TGFβ signaling with a small-molecule inhibitor or a monoclonal antibody did not improve survival. Conversely, tumor antigen-specific vaccination in combination with focal radiation therapy reversed tolerance and improved survival. This treatment regimen was associated with increased polyfunctionality of CD8 T cells, elevated T effector to T regulatory cell ratios, and decreased TGFβ secretion from microglia. CONCLUSIONS These data suggest that CNS tumors may impair systemic antitumor immunity and consequently accelerate cancer progression locally as well as outside the CNS, whereas antitumor immunity may be restored by combining vaccination with radiation therapy. These findings are hypothesis-generating and warrant further study in contemporary melanoma models as well as human trials.
Collapse
Affiliation(s)
| | | | | | - Nicholas M Durham
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Jacob Ruzevick
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Angela Alme
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Brian J Francica
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Jimmy Elias
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Andrew Daniels
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | | | | | | | - Emily G Baxi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland. Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland. Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland. Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland. Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland.
| | - Charles G Drake
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
17
|
WANG YONGGANG, ZHAN YIPING, PAN SHUYI, WANG HAIDONG, ZHANG DUNXIAO, GAO KAI, QI XUELING, YU CHUNJIANG. Hyperbaric oxygen promotes malignant glioma cell growth and inhibits cell apoptosis. Oncol Lett 2015; 10:189-195. [PMID: 26170997 PMCID: PMC4487135 DOI: 10.3892/ol.2015.3244] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 04/13/2015] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequently diagnosed intracranial malignant tumor in adults. Clinical studies have indicated that hyperbaric oxygen may improve the prognosis and reduce complications in glioma patients; however, the specific mechanism by which this occurs remains unknown. The present study investigated the direct effects of hyperbaric oxygen stimulation on glioma by constructing an intracranial transplanted glioma model in congenic C57BL/6J mice. Bioluminescent imaging (BLI) was used to assess the growth of intracranial transplanted GL261-Luc glioma cells in vivo, while flow cytometric and immunohistochemical assays were used to detect and compare the expression of the biomarkers, Ki-67, CD34 and TUNEL, reflecting the cell cycle, apoptosis and angiogenesis. BLI demonstrated that hyperbaric oxygen promoted the growth of intracranially transplanted GL261-Luc glioma cells in vivo. Flow cytometric analysis indicated that hyperbaric oxygen promoted GL261-Luc glioma cell proliferation and also prevented cell cycle arrest. In addition, hyperbaric oxygen inhibited the apoptosis of the transplanted glioma cells. Immunohistochemical analysis also indicated that hyperbaric oxygen increased positive staining for Ki-67 and CD34, while reducing staining for TUNEL (a marker of apoptosis). The microvessel density was significantly increased in the hyperbaric oxygen treatment group compared with the control group. In conclusion, hyperbaric oxygen treatment promoted the growth of transplanted malignant glioma cells in vivo and also inhibited the apoptosis of these cells.
Collapse
Affiliation(s)
- YONG-GANG WANG
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - YI-PING ZHAN
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - SHU-YI PAN
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100048, P.R. China
| | - HAI-DONG WANG
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100048, P.R. China
| | - DUN-XIAO ZHANG
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100048, P.R. China
| | - KAI GAO
- Institute of Laboratory Animal Sciences, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - XUE-LING QI
- Department of Pathology, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - CHUN-JIANG YU
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| |
Collapse
|
18
|
Morin-Brureau M, Hooper KM, Prosniak M, Sauma S, Harshyne LA, Andrews DW, Hooper DC. Enhancement of glioma-specific immunity in mice by "NOBEL", an insulin-like growth factor 1 receptor antisense oligodeoxynucleotide. Cancer Immunol Immunother 2015; 64:447-57. [PMID: 25579379 PMCID: PMC11028597 DOI: 10.1007/s00262-015-1654-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/03/2015] [Indexed: 10/24/2022]
Abstract
Autologous glioblastoma multiforme tumor cells treated with an antisense oligodeoxynucleotide (AS-ODN) targeting insulin-like growth factor receptor-1 (IGF-1R) are the basis of a vaccine with therapeutic effects on tumor recurrence in a pilot clinical trial. As a preface to continued clinical investigation of this vaccination strategy, we have studied the contribution of an optimized IGF-1R AS-ODN, designated "NOBEL", to the induction of immunity to mouse GL261 glioma cells. The impact of NOBEL on mechanisms contributing to the development of GL261 immunity was first examined in the periphery. GL261 cells are naturally immunogenic when implanted into the flanks of congenic C57BL/6 mice, immunizing rather than forming tumors in around 50 % of these animals but causing tumors in the majority of mice lacking T and B lymphocytes. Overnight treatment with NOBEL in vitro reduces IGF-1R expression by GL261 cells but has minimal effect on cell viability and does not reduce the capacity of the cells to form tumors upon implantation. In contrast, tumors are extremely rare when GL261 cells are mixed with NOBEL at inoculation into the flanks of C57BL/6, and the recipient mice become immune to subcutaneous and intracranial challenge with untreated GL261. Adaptive immune mechanisms contribute to this effect, as immunocompromised mice fail to either fully control tumor formation or develop immunity following flank administration of the GL261/NOBEL mix. NOBEL's structure has known immunostimulatory motifs that likely contribute to the immunogenicity of the mix, but its specificity for IGF-1R mRNA is also important as a similarly structured sense molecule is not effective.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Brain Neoplasms/immunology
- Brain Neoplasms/pathology
- Brain Neoplasms/therapy
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Glioma/immunology
- Glioma/pathology
- Glioma/therapy
- Immunity, Cellular/immunology
- Immunotherapy
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oligodeoxyribonucleotides, Antisense/administration & dosage
- Oligodeoxyribonucleotides, Antisense/genetics
- Oligodeoxyribonucleotides, Antisense/immunology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- beta 2-Microglobulin/physiology
Collapse
Affiliation(s)
- Mélanie Morin-Brureau
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Kirsten M. Hooper
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107 USA
- Present Address: Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA USA
| | - Michael Prosniak
- Department of Cancer Biology, Thomas Jefferson University, 1020 Locust Street, JAH Rm 452, Philadelphia, PA 19107-6731 USA
| | - Sami Sauma
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Larry A. Harshyne
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - David W. Andrews
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - D. Craig Hooper
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA 19107 USA
- Department of Cancer Biology, Thomas Jefferson University, 1020 Locust Street, JAH Rm 452, Philadelphia, PA 19107-6731 USA
| |
Collapse
|
19
|
Feng E, Gao H, Su W, Yu C. Immunotherapy of rat glioma without accumulation of CD4(+)CD25(+)FOXP3(+) regulatory T cells. Neural Regen Res 2015; 7:1498-506. [PMID: 25657686 PMCID: PMC4308782 DOI: 10.3969/j.issn.1673-5374.2012.19.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 05/18/2012] [Indexed: 11/18/2022] Open
Abstract
Immunotherapy may be used for the treatment of glioblastoma multiforme; however, the induced immune response is inadequate when either T cells or dendritic cells are used alone. In this study, we established a novel vaccine procedure in rats, using dendritic cells pulsed with C6 tumor cell lysates in combination with adoptive transfer of T lymphocytes from syngenic donors. On day 21 after tumor inoculation, all the rats were sacrificed, the brains were harvested for calculation of glioma volume, cytolytic T lymphocyte responses were measured by cytotoxic assay, and the frequency of regulatory T lymphocytes (CD4+CD25+FOXP3+) in the peripheral blood was investigated by flow cytometric analysis. The survival rate of rats bearing C6 glioma was observed. Results showed that the co-immunization strategy had significant anti-tumor potential against the pre-established C6 glioma, and induced a strong cytolytic T lymphocyte response in rats. The frequency of peripheral blood CD4+CD25+FOXP3+ regulatory T lymphocytes was significantly decreased following the combination therapy, and the rats survived for a longer period. Experimental findings indicate that the combined immunotherapy of glioma cell lysate-pulsed dendritic cell vaccination following adoptive transfer of T cells can effectively inhibit the growth of gliomas in rats, boost anti-tumor immunity and produce a sustained immune response while avoiding the accumulation of CD4+CD25+FOXP3+ regulatory T lymphocytes.
Collapse
Affiliation(s)
- Enshan Feng
- Fuxing Hospital, Capital Medical University, Beijing 100038, China
| | - Haili Gao
- Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Wei Su
- Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Chunjiang Yu
- Fuxing Hospital, Capital Medical University, Beijing 100038, China
| |
Collapse
|
20
|
Clark AJ, Safaee M, Oh T, Ivan ME, Parimi V, Hashizume R, Ozawa T, James CD, Bloch O, Parsa AT. Stable luciferase expression does not alter immunologic or in vivo growth properties of GL261 murine glioma cells. J Transl Med 2014; 12:345. [PMID: 25464980 PMCID: PMC4258256 DOI: 10.1186/s12967-014-0345-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/24/2014] [Indexed: 11/17/2022] Open
Abstract
Background GL261 cells are murine glioma cells that demonstrate proliferation, invasion, and angiogenesis when implanted in syngeneic C57BL/6 mice, providing a highly useful immunocompetent animal model of glioblastoma. Modification of tumor cells for luciferase expression enables non-invasive monitoring of orthotopic tumor growth, and has proven useful for studying glioblastoma response to novel therapeutics. However, tumor modification for luciferase has the potential for evoking host immune response against otherwise syngeneic tumor cells, thereby mitigating the tumor cells’ value for tumor immunology and immunotherapy studies. Methods GL261 cells were infected with lentivirus containing a gene encoding firefly luciferase (GL261.luc). In vitro proliferation of parental (unmodified) GL261 and GL261.luc was measured on days 0, 1, 2, 4, and 7 following plating, and the expression of 82 mouse cytokines and chemokines were analyzed by RT-PCR array. Cell lines were also evaluated for differences in invasion and migration in modified Boyden chambers. GL261 and GL261.luc cells were then implanted intracranially in C57BL/6 mice, with GL261.luc tumor growth monitored by quantitative bioluminescence imaging, and all mice were followed for survival to compare relative malignancy of tumor cells. Results No difference in proliferation was indicated for GL261 vs. GL261.luc cells (p>0.05). Of the 82 genes examined by RT-PCR array, seven (9%) exhibited statistically significant change after luciferase modification. Of these, only three changed by greater than 2-fold: BMP-2, IL-13, and TGF-β2. No difference in invasion (p=0.67) or migration (p=0.26) was evident between modified vs. unmodified cells. GL261.luc cell luminescence was detectable in the brains of C57BL/6 mice at day 5 post-implantation, and tumor bioluminescence increased exponentially to day 19. Median overall survival was 20.2 days versus 19.7 days for mice receiving implantation with GL261 and GL261.luc, respectively (p=0.62). Histopathologic analysis revealed no morphological difference between tumors, and immunohistochemical analysis showed no significant difference for staining of CD3, Ki67, or CD31 (p>0.05 for all). Conclusions Luciferase expression in GL261 murine glioma cells does not affect GL261 proliferation, invasion, cytokine expression, or in vivo growth. Luciferase modification increases their utility for studying tumor immunology and immunotherapeutic approaches for treating glioblastoma.
Collapse
Affiliation(s)
- Aaron J Clark
- The Brain Tumor Research Center, Department of Neurological Surgery, University of California, San Francisco, CA 505 Parnassus Ave., Room 779 M, San Francisco, CA, 94143-0112, USA. .,Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Ave., M779, Box 0112, San Francisco, CA, 94117, USA.
| | - Michael Safaee
- The Brain Tumor Research Center, Department of Neurological Surgery, University of California, San Francisco, CA 505 Parnassus Ave., Room 779 M, San Francisco, CA, 94143-0112, USA.
| | - Taemin Oh
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair St., Suite 2210, Chicago, IL, 60611-2922, USA.
| | - Michael E Ivan
- The Brain Tumor Research Center, Department of Neurological Surgery, University of California, San Francisco, CA 505 Parnassus Ave., Room 779 M, San Francisco, CA, 94143-0112, USA.
| | - Vamsi Parimi
- Pathology Core Facility, Feinberg School of Medicine, Northwestern University, 710 N. Fairbanks Court, Room 8-419, Chicago, IL, USA.
| | - Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair St., Suite 2210, Chicago, IL, 60611-2922, USA.
| | - Tomoko Ozawa
- The Brain Tumor Research Center, Department of Neurological Surgery, University of California, San Francisco, CA 505 Parnassus Ave., Room 779 M, San Francisco, CA, 94143-0112, USA.
| | - Charles D James
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair St., Suite 2210, Chicago, IL, 60611-2922, USA.
| | - Orin Bloch
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair St., Suite 2210, Chicago, IL, 60611-2922, USA.
| | - Andrew T Parsa
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, 676 N. St. Clair St., Suite 2210, Chicago, IL, 60611-2922, USA.
| |
Collapse
|
21
|
Tucci M, Stucci S, Passarelli A, Giudice G, Dammacco F, Silvestris F. The immune escape in melanoma: role of the impaired dendritic cell function. Expert Rev Clin Immunol 2014; 10:1395-404. [DOI: 10.1586/1744666x.2014.955851] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
22
|
Xu M, Yao Y, Hua W, Wu Z, Zhong P, Mao Y, Zhou L, Luo F, Chu Y. Mouse glioma immunotherapy mediated by A2B5+ GL261 cell lysate-pulsed dendritic cells. J Neurooncol 2014; 116:497-504. [DOI: 10.1007/s11060-013-1334-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 12/27/2013] [Indexed: 10/25/2022]
|
23
|
Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H. The brain tumor microenvironment. Glia 2013; 60:502-14. [PMID: 22379614 DOI: 10.1002/glia.21264] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
High-grade brain tumors are heterogeneous with respect to the composition of bona fide tumor cells and with respect to a range of intermingling parenchymal cells. Glioblastomas harbor multiple cell types, some with increased tumorigenicity and stem cell-like capacity. The stem-like cells maybe the cells of origin for tumor relapse. However, the tumor-associated parenchymal cells such as vascular cells,microglia, peripheral immune cells, and neural precursor cells also play a vital role in controlling the course of pathology.In this review, we describe the multiple interactions of bulk glioma cells and glioma stem cells with parenchymal cell populations and highlight the pathological impact as well as signaling pathways known for these types of cell-cell communication. The tumor-vasculature not only nourishes glioblastomas, but also provides a specialized niche for these stem-like cells. In addition, microglial cells,which can contribute up to 30% of a brain tumor mass,play a role in glioblastoma cell invasion. Moreover, non-neoplastic astrocytes can be converted into a reactive phenotype by the glioma microenvironment and can then secrete a number of factors which influences tumor biology. The young brain may have the capacity to inhibit gliomagenesis by the endogenous neural precursor cells, which secrete tumor suppressive factors. The factors, pathways, and interactions described in this review provide a new prospective on the cell biology of primary brain tumors, which may ultimately generate new treatment modalities. However, our picture of the multiple interactions between parenchymal and tumor cells is still incomplete.
Collapse
Affiliation(s)
- Nikki A Charles
- Brain Tumor Center and Department of Neurosurgery, Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | |
Collapse
|
24
|
Goffart N, Kroonen J, Rogister B. Glioblastoma-initiating cells: relationship with neural stem cells and the micro-environment. Cancers (Basel) 2013; 5:1049-71. [PMID: 24202333 PMCID: PMC3795378 DOI: 10.3390/cancers5031049] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 07/29/2013] [Accepted: 08/01/2013] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma multiforme (GBM, WHO grade IV) is the most common and lethal subtype of primary brain tumor with a median overall survival of 15 months from the time of diagnosis. The presence in GBM of a cancer population displaying neural stem cell (NSC) properties as well as tumor-initiating abilities and resistance to current therapies suggests that these glioblastoma-initiating cells (GICs) play a central role in tumor development and are closely related to NSCs. However, it is nowadays still unclear whether GICs derive from NSCs, neural progenitor cells or differentiated cells such as astrocytes or oligodendrocytes. On the other hand, NSCs are located in specific regions of the adult brain called neurogenic niches that have been shown to control critical stem cell properties, to nourish NSCs and to support their self-renewal. This “seed-and-soil” relationship has also been adapted to cancer stem cell research as GICs also require a specific micro-environment to maintain their “stem cell” properties. In this review, we will discuss the controversies surrounding the origin and the identification of GBM stem cells and highlight the micro-environment impact on their biology.
Collapse
Affiliation(s)
- Nicolas Goffart
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences Research Center, University of Liège, Liège 4000, Belgium; E-Mail:
| | - Jérôme Kroonen
- Human Genetics, CHU and University of Liège, Liège 4000, Belgium; E-Mail:
- The T&P Bohnenn Laboratory for Neuro-Oncology, Department of Neurology and Neurosurgery, UMC Utrecht, Utrecht 3556, The Netherlands; E-Mail:
| | - Bernard Rogister
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences Research Center, University of Liège, Liège 4000, Belgium; E-Mail:
- Department of Neurology, CHU and University of Liège, Liège 4000, Belgium
- GIGA-Development, Stem Cells and Regenerative Medicine, University of Liège, Liège 4000, Belgium
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +32-4-366-5950; Fax: +32-4-366-5912
| |
Collapse
|
25
|
Foulds GA, Radons J, Kreuzer M, Multhoff G, Pockley AG. Influence of tumors on protective anti-tumor immunity and the effects of irradiation. Front Oncol 2013; 3:14. [PMID: 23378947 PMCID: PMC3561630 DOI: 10.3389/fonc.2013.00014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/15/2013] [Indexed: 12/20/2022] Open
Abstract
Innate and adaptive immunity plays important roles in the development and progression of cancer and it is becoming apparent that tumors can influence the induction of potentially protective responses in a number of ways. The prevalence of immunoregulatory T cell populations in the circulation and tumors of patients with cancer is increased and the presence of these cells appears to present a major barrier to the induction of tumor immunity. One aspect of tumor-mediated immunoregulation which has received comparatively little attention is that which is directed toward natural killer (NK) cells, although evidence that the phenotype and function of NK cell populations are modified in patients with cancer is accumulating. Although the precise mechanisms underlying these localized and systemic immunoregulatory effects remain unclear, tumor-derived factors appear, in part at least, to be involved. The effects could be manifested by an altered function and/or via an influence on the migratory properties of individual cell subsets. A better insight into endogenous immunoregulatory mechanisms and the capacity of tumors to modify the phenotype and function of innate and adaptive immune cells might assist the development of new immunotherapeutic approaches and improve the management of patients with cancer. This article reviews current knowledge relating to the influence of tumors on protective anti-tumor immunity and considers the potential influence that radiation-induced effects might have on the prevalence, phenotype, and function of innate and adaptive immune cells in patients with cancer.
Collapse
Affiliation(s)
- Gemma A Foulds
- Department of Oncology, The Medical School, The University of Sheffield Sheffield, UK ; Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München Munich, Germany
| | | | | | | | | |
Collapse
|
26
|
Abstract
The brain is in many ways an immunologically and pharmacologically privileged site. The blood-brain barrier (BBB) of the cerebrovascular endothelium and its participation in the complex structure of the neurovascular unit (NVU) restrict access of immune cells and immune mediators to the central nervous system (CNS). In pathologic conditions, very well-organized immunologic responses can develop within the CNS, raising important questions about the real nature and the intrinsic and extrinsic regulation of this immune privilege. We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors. The goals of this review are to outline key scientific advances and the status of the science central to both the neuroinflammation and CNS barriers fields, and highlight the opportunities and priorities in advancing brain barriers research in the context of the larger immunology and neuroscience disciplines. This review article was developed from reports presented at the 2011 Annual Blood-Brain Barrier Consortium Meeting.
Collapse
|
27
|
Abstract
Indoleamine 2,3-dioxygenase (IDO) is an enzyme with known immunosuppressive and tolerogenic effects in cancer. Mounting evidence has associated IDO expression with the induction of regulatory T cells (Treg) and malignant progression. IDO inhibition may therefore provide a promising therapeutic approach for glioblastoma, where the need for novel treatment is great.
Collapse
Affiliation(s)
- Bryan D Choi
- Duke Brain Tumor Immunotherapy Program, Preston Robert Tisch Brain Tumor Center, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
28
|
|
29
|
Donnou S, Galand C, Daussy C, Crozet L, Fridman WH, Sautès-Fridman C, Fisson S. Immune adaptive microenvironment profiles in intracerebral and intrasplenic lymphomas share common characteristics. Clin Exp Immunol 2011; 165:329-37. [PMID: 21668435 DOI: 10.1111/j.1365-2249.2011.04416.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A large body of evidence indicates that the immune microenvironment controls tumour development. Primary central nervous system lymphomas (PCNSL) are aggressive tumours growing in the central nervous system (CNS). To evaluate the role and characteristics of this immune-privileged site in anti-tumour defences, we compared the cellular and molecular immune microenvironments of growing murine lymphoma B cells injected into the brain or the spleen. In the brain, immune cells, including dendritic cells and T lymphocytes with a large proportion of CD4(+) forkhead box P3 (FoxP3(+)) regulatory T cells, rapidly infiltrated the tumour microenvironment. These populations also increased in number in the spleen. The T cell cytokine profiles in tumour-bearing mice were similar in the two sites, with predominant T helper type 1 (Th1)/Th17 polarization after polyclonal stimulation, although some interleukin (IL)-4 could also be found. We demonstrated that these T cells have anti-tumour activity in the CNS, although less than in the spleen: nude mice that received lymphoma cells intracerebrally died significantly earlier than immunocompetent animals. These results demonstrate that the brain is able to recruit all the major actors to mount a specific anti-tumour immune response against lymphoma.
Collapse
Affiliation(s)
- S Donnou
- Institut National de Santé et de Recherche Médicale (INSERM), UMRS872, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | | | | | | |
Collapse
|
30
|
Ataera H, Hyde E, Price KM, Stoitzner P, Ronchese F. Murine melanoma-infiltrating dendritic cells are defective in antigen presenting function regardless of the presence of CD4CD25 regulatory T cells. PLoS One 2011; 6:e17515. [PMID: 21390236 PMCID: PMC3048402 DOI: 10.1371/journal.pone.0017515] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 02/04/2011] [Indexed: 01/08/2023] Open
Abstract
Tumor-infiltrating dendritic cells are often ineffective at presenting tumor-derived antigen in vivo, a defect usually ascribed to the suppressive tumor environment. We investigated the effects of depleting CD4+CD25+ “natural” regulatory T cells (Treg) on the frequency, phenotype and function of total dendritic cell populations in B16.OVA tumors and in tumor-draining lymph nodes. Intraperitoneal injection of the anti-CD25 monoclonal antibody PC61 reduced Treg frequency in blood and tumors, but did not affect the frequency of tumor-infiltrating dendritic cells, or their expression of CD40, CD86 and MHCII. Tumor-infiltrating dendritic cells from PC61-treated or untreated mice induced the proliferation of allogeneic T cells in vitro, but could not induce proliferation of OVA-specific OTI and OTII T cells unless specific peptide antigen was added in culture. Some proliferation of naïve, OVA-specific OTI T cells, but not OTII T cells, was observed in the tumor-draining LN of mice carrying B16.OVA tumors, however, this was not improved by PC61 treatment. Experiments using RAG1−/− hosts adoptively transferred with OTI and CD25-depleted OTII cells also failed to show improved OTI and OTII T cell proliferation in vivo compared to C57BL/6 hosts. We conclude that the defective presentation of B16.OVA tumor antigen by tumor-infiltrating dendritic cells and in the tumor-draining lymph node is not due to the presence of “natural” CD4+CD25+ Treg.
Collapse
Affiliation(s)
- Haley Ataera
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Evelyn Hyde
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Kylie M. Price
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
- * E-mail:
| |
Collapse
|
31
|
Wei J, Wu A, Kong LY, Wang Y, Fuller G, Fokt I, Melillo G, Priebe W, Heimberger AB. Hypoxia potentiates glioma-mediated immunosuppression. PLoS One 2011; 6:e16195. [PMID: 21283755 PMCID: PMC3024401 DOI: 10.1371/journal.pone.0016195] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 12/14/2010] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a lethal cancer that exerts potent immune suppression. Hypoxia is a predominant feature of GBM, but it is unclear to the degree in which tumor hypoxia contributes to this tumor-mediated immunosuppression. Utilizing GBM associated cancer stem cells (gCSCs) as a treatment resistant population that has been shown to inhibit both innate and adaptive immune responses, we compared immunosuppressive properties under both normoxic and hypoxic conditions. Functional immunosuppression was characterized based on production of immunosuppressive cytokines and chemokines, the inhibition of T cell proliferation and effector responses, induction of FoxP3+ regulatory T cells, effect on macrophage phagocytosis, and skewing to the immunosuppressive M2 phenotype. We found that hypoxia potentiated the gCSC-mediated inhibition of T cell proliferation and activation and especially the induction of FoxP3+T cells, and further inhibited macrophage phagocytosis compared to normoxia condition. These immunosuppressive hypoxic effects were mediated by signal transducer and activator of transcription 3 (STAT3) and its transcriptionally regulated products such as hypoxia inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF). Inhibitors of STAT3 and HIF-1α down modulated the gCSCs' hypoxia-induced immunosuppressive effects. Thus, hypoxia further enhances GBM-mediated immunosuppression, which can be reversed with therapeutic inhibition of STAT3 and HIF-1α and also helps to reconcile the disparate findings that immune therapeutic approaches can be used successfully in model systems but have yet to achieve generalized successful responses in the vast majority of GBM patients by demonstrating the importance of the tumor hypoxic environment.
Collapse
Affiliation(s)
- Jun Wei
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Adam Wu
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Yongtao Wang
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Gregory Fuller
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Isabella Fokt
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Giovanni Melillo
- National Cancer Institute, Frederick, Maryland, United States of America
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Amy B. Heimberger
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Wu A, Wei J, Kong LY, Wang Y, Priebe W, Qiao W, Sawaya R, Heimberger AB. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol 2010; 12:1113-25. [PMID: 20667896 PMCID: PMC3098021 DOI: 10.1093/neuonc/noq082] [Citation(s) in RCA: 495] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 06/15/2010] [Indexed: 02/07/2023] Open
Abstract
Macrophages (MΦs)/microglia that constitute the dominant tumor-infiltrating immune cells in glioblastoma are recruited by tumor-secreted factors and are induced to become immunosuppressive and tumor supportive (M2). Glioma cancer stem cells (gCSCs) have been shown to suppress adaptive immunity, but their role in innate immunity with respect to the recruitment and polarization of MΦs/microglia is unknown. The innate immunosuppressive properties of the gCSCs were characterized based on elaborated MΦ inhibitory cytokine-1 (MIC-1), transforming growth factor (TGF-β1), soluble colony-stimulating factor (sCSF), recruitment of monocytes, inhibition of MΦ/microglia phagocytosis, induction of MΦ/microglia cytokine secretion, and the inhibition of T-cell proliferation. The role of the signal transducer and activator of transcription 3 (STAT3) in mediating innate immune suppression was evaluated in the context of the functional assays. The gCSCs produced sCSF-1, TGF-β1, and MIC-1, cytokines known to recruit and polarize the MΦs/microglia to become immunosuppressive. The gCSC-conditioned medium polarized the MΦ/microglia to an M2 phenotype, inhibited MΦ/microglia phagocytosis, induced the secretion of the immunosuppressive cytokines interleukin-10 (IL-10) and TGF-β1 by the MΦs/microglia, and enhanced the capacity of MΦs/microglia to inhibit T-cell proliferation. The inhibition of phagocytosis and the secretion of IL-10 were reversed when the STAT3 pathway was blocked in the gCSCs. The gCSCs modulate innate immunity in glioblastoma by inducing immunosuppressive MΦs/microglia, and this capacity can be reversed by inhibiting phosphorylated STAT3.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Amy B. Heimberger
- Department of Neurosurgery (A.W., J.W., L.-Y.K., Y.W., R.S., A.B.H.), Department of Experimental Therapeutics (W.P.), Department of Biostatistics (W.Q.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
33
|
Tran Thang NN, Derouazi M, Philippin G, Arcidiaco S, Di Berardino-Besson W, Masson F, Hoepner S, Riccadonna C, Burkhardt K, Guha A, Dietrich PY, Walker PR. Immune infiltration of spontaneous mouse astrocytomas is dominated by immunosuppressive cells from early stages of tumor development. Cancer Res 2010; 70:4829-39. [PMID: 20501837 DOI: 10.1158/0008-5472.can-09-3074] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immune infiltration of advanced human gliomas has been shown, but it is doubtful whether these immune cells affect tumor progression. It could be hypothesized that this infiltrate reflects recently recruited immune cells that are immediately overwhelmed by a high tumor burden. Alternatively, if there is earlier immune detection and infiltration of the tumor, the question arises as to when antitumor competency is lost. To address these issues, we analyzed a transgenic mouse model of spontaneous astrocytoma (GFAP-V(12)HA-ras mice), which allows the study of immune interactions with developing glioma, even at early asymptomatic stages. T cells, including a significant proportion of Tregs, are already present in the brain before symptoms develop, followed later by macrophages, natural killer cells, and dendritic cells. The effector potential of CD8 T-cells is defective, with the absence of granzyme B expression and low expression of IFN-gamma, tumor necrosis factor, and interleukin 2. Overall, our results show an early defective endogenous immune response to gliomas, and local accumulation of immunosuppressive cells at the tumor site. Thus, the antiglioma response is not simply overwhelmed at advanced stages of tumor growth, but is counterbalanced by an inhibitory microenvironment from the outset. Nevertheless, we determined that effector molecule expression (granzyme B, IFN-gamma) by brain-infiltrating CD8 T-cells could be enhanced, despite this unfavorable milieu, by strong immune stimuli. This potential to modulate the strong imbalance in local antiglioma immunity is encouraging for the development and optimization of future glioma immunotherapies.
Collapse
|
34
|
Hau P, Jachimczak P, Bogdahn U. Treatment of malignant gliomas with TGF-beta2 antisense oligonucleotides. Expert Rev Anticancer Ther 2010; 9:1663-74. [PMID: 19895249 DOI: 10.1586/era.09.138] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Antisense oligodeoxynucleotides (AS-ODNs) have been widely used to determine gene function, validate drug targets and as novel therapeutics for human diseases. In this review, we describe the development of AS-ODNs, including their modifications, pharmacokinetics and toxicity in animal models and humans, and their preclinical and clinical development in the therapy of human high-grade gliomas. The most advanced AS-ODN for the therapy of high-grade gliomas is a phosphorothioate-modified AS-ODN, AP 12009 (trabedersen), which targets mRNA encoding TGF-beta2. AP 12009 is administered intratumorally using convection-enhanced delivery. A series of Phase I and II clinical trials have evaluated the toxicity profile and optimal dose of the substance. A randomized, controlled international Phase III study was initiated in March 2009 and will compare trabedersen 10 microM versus conventional alkylating chemotherapy in patients with recurrent or refractory anaplastic astrocytoma after standard radio- and chemotherapy.
Collapse
Affiliation(s)
- Peter Hau
- Department of Neurology, University of Regensburg, Medical School, Regensburg, Germany.
| | | | | |
Collapse
|