151
|
de Groot J, Penas-Prado M, Alfaro-Munoz K, Hunter K, Pei BL, O'Brien B, Weathers SP, Loghin M, Kamiya Matsouka C, Yung WKA, Mandel J, Wu J, Yuan Y, Zhou S, Fuller GN, Huse J, Rao G, Weinberg JS, Prabhu SS, McCutcheon IE, Lang FF, Ferguson SD, Sawaya R, Colen R, Yadav SS, Blando J, Vence L, Allison J, Sharma P, Heimberger AB. Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages. Neuro Oncol 2021; 22:539-549. [PMID: 31755915 DOI: 10.1093/neuonc/noz185] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We sought to ascertain the immune effector function of pembrolizumab within the glioblastoma (GBM) microenvironment during the therapeutic window. METHODS In an open-label, single-center, single-arm phase II "window-of-opportunity" trial in 15 patients with recurrent (operable) GBM receiving up to 2 pembrolizumab doses before surgery and every 3 weeks afterward until disease progression or unacceptable toxicities occurred, immune responses were evaluated within the tumor. RESULTS No treatment-related deaths occurred. Overall median follow-up time was 50 months. Of 14 patients monitored, 10 had progressive disease, 3 had a partial response, and 1 had stable disease. Median progression-free survival (PFS) was 4.5 months (95% CI: 2.27, 6.83), and the 6-month PFS rate was 40%. Median overall survival (OS) was 20 months, with an estimated 1-year OS rate of 63%. GBM patients' recurrent tumors contained few T cells that demonstrated a paucity of immune activation markers, but the tumor microenvironment was markedly enriched for CD68+ macrophages. CONCLUSIONS Immune analyses indicated that pembrolizumab anti-programmed cell death 1 (PD-1) monotherapy alone can't induce effector immunologic response in most GBM patients, probably owing to a scarcity of T cells within the tumor microenvironment and a CD68+ macrophage preponderance.
Collapse
Affiliation(s)
- John de Groot
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marta Penas-Prado
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kristin Alfaro-Munoz
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathy Hunter
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Be Lian Pei
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara O'Brien
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shiao-Pei Weathers
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monica Loghin
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carlos Kamiya Matsouka
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - W K Alfred Yung
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jacob Mandel
- Department of Neurology, Baylor College of Medicine, Houston, Texas
| | - Jimin Wu
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ying Yuan
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shouhao Zhou
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Huse
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ganesh Rao
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey S Weinberg
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sujit S Prabhu
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ian E McCutcheon
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F Lang
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherise D Ferguson
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raymond Sawaya
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rivka Colen
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shalini S Yadav
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luis Vence
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James Allison
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Padmanee Sharma
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
152
|
Abstract
BACKGROUND Vestibular schwannomas exhibit a uniquely variable natural history of growth, stability, or even spontaneous regression. We hypothesized that a transitory population of immune cells, or immunomodulation of tumors cells, may influence the growth pattern of schwannomas. We therefore sought to characterize the impact of the immune microenvironment on schwannoma behavior. METHODS Forty-eight vestibular schwannomas with preoperative magnetic resonance imaging and 11 with serial imaging were evaluated for presence of immune infiltrates (including the pan-leukocyte marker Cluster of Differentiation (CD)45, CD4 and CD8 T-cell, and CD68 and CD163 macrophages) as well as expression of immunomodulatory regulators (Programmed Death Ligand 1 (PD-L1), Programmed Death Ligand 2 (PD-L2), LAG-3, TIM-3, V-domain Ig Suppressor of T cell Activation). Maximal diameter, volume, and recurrence were annotated. RESULTS Vestibular schwannomas were characterized by diverse signatures of tumor infiltrating leukocytes and immunomodulatory markers. The median tumor volume was 4.7 cm (Interquartile Range (IQR) 1.0-13.0) and maximum diameter was 2.3 cm (IQR 1.5-3.2). Among tumors with serial imaging, the median volumetric growth was 0.04 cm/mo (IQR 0.01-0.18). Tumor volume and maximum diameter demonstrated strong concordance (R = 0.90; p < 0.001). Vestibular schwannoma volume was positively associated with CD4, CD68, and CD163, but not CD8, immune infiltration (all p < 0.05). Tumor growth was positively associated with CD163 and PD-L1 (both p < 0.05). Further, CD163 modified this effect: the relationship between PD-L1 and growth strengthened with increasing CD163 infiltration (R = 0.81, p = 0.007). No other immune cell types modified this relationship. These associations were inconsistently observed for maximum diameter and linear growth. CONCLUSION Vestibular schwannomas demonstrate variable expression of immune regulatory markers as well as immune infiltrates. Tumor size is associated with immune infiltrates and tumor growth is associated with PD-L1, especially in the presence of M2-subtype macrophages. Volumetric measures may associate with the biological signature more accurately than linear parameters. Future exploration of the role of immune modulation in select schwannomas will further enhance our understanding of the biology of these tumors and suggest potential therapeutic avenues for control of tumor growth.
Collapse
|
153
|
Wang Z, Wang Y, Yang T, Xing H, Wang Y, Gao L, Guo X, Xing B, Wang Y, Ma W. Machine learning revealed stemness features and a novel stemness-based classification with appealing implications in discriminating the prognosis, immunotherapy and temozolomide responses of 906 glioblastoma patients. Brief Bioinform 2021; 22:6220175. [PMID: 33839757 PMCID: PMC8425448 DOI: 10.1093/bib/bbab032] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant and lethal intracranial tumor, with extremely limited treatment options. Immunotherapy has been widely studied in GBM, but none can significantly prolong the overall survival (OS) of patients without selection. Considering that GBM cancer stem cells (CSCs) play a non-negligible role in tumorigenesis and chemoradiotherapy resistance, we proposed a novel stemness-based classification of GBM and screened out certain population more responsive to immunotherapy. The one-class logistic regression algorithm was used to calculate the stemness index (mRNAsi) of 518 GBM patients from The Cancer Genome Atlas (TCGA) database based on transcriptomics of GBM and pluripotent stem cells. Based on their stemness signature, GBM patients were divided into two subtypes via consensus clustering, and patients in Stemness Subtype I presented significantly better OS but poorer progression-free survival than Stemness Subtype II. Genomic variations revealed patients in Stemness Subtype I had higher somatic mutation loads and copy number alteration burdens. Additionally, two stemness subtypes had distinct tumor immune microenvironment patterns. Tumor Immune Dysfunction and Exclusion and subclass mapping analysis further demonstrated patients in Stemness Subtype I were more likely to respond to immunotherapy, especially anti-PD1 treatment. The pRRophetic algorithm also indicated patients in Stemness Subtype I were more resistant to temozolomide therapy. Finally, multiple machine learning algorithms were used to develop a 7-gene Stemness Subtype Predictor, which were further validated in two external independent GBM cohorts. This novel stemness-based classification could provide a promising prognostic predictor for GBM and may guide physicians in selecting potential responders for preferential use of immunotherapy.
Collapse
Affiliation(s)
- Zihao Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yaning Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tianrui Yang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hao Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yuekun Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lu Gao
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiaopeng Guo
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Bing Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
154
|
Lanza M, Casili G, Campolo M, Paterniti I, Colarossi C, Mare M, Giuffrida R, Caffo M, Esposito E, Cuzzocrea S. Immunomodulatory Effect of Microglia-Released Cytokines in Gliomas. Brain Sci 2021; 11:brainsci11040466. [PMID: 33917013 PMCID: PMC8067679 DOI: 10.3390/brainsci11040466] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/16/2021] [Accepted: 04/02/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia, a type of differentiated tissue macrophage, are considered to be the most plastic cell population of the central nervous system (CNS). Microglia substantially contribute to the growth and invasion of tumor mass in brain tumors including glioblastoma (GB). In response to pathological conditions, resting microglia undergo a stereotypic activation process and become capable of phagocytosis, antigen presentation, and lymphocyte activation. Considering their immune effector function, it is not surprising to see microglia accumulation in almost every CNS disease process, including malignant brain tumors. Large numbers of glioma associated microglia and macrophages (GAMs) can accumulate within the tumor where they appear to have an important role in prognosis. GAMs constitute the largest portion of tumor infiltrating cells, contributing up to 30% of the entire glioma mass and upon interaction with neoplastic cells. GAMs acquire a unique phenotype of activation, including both M1 and M2 specific markers. It has been demonstrated that microglia possess a dual role: on one hand, microglia may represent a CNS anti-tumor response, which is inactivated by local secretion of immunosuppressive factors by glioma cells. On the other hand, taking into account that microglia are capable of secreting a variety of immunomodulatory cytokines, it is possible that they are attracted by gliomas to promote tumor growth. A better understanding of microglia-glioma interaction will be helpful in designing novel immune-based therapies against these fatal tumors. Concluding, as microglia significantly may contribute to glioma biology, favoring tumor growth and invasiveness, these cells represent a valuable alternative/additional target for the development of more effective treatments for gliomas.
Collapse
Affiliation(s)
- Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98166 Messina, Italy; (M.L.); (G.C.); (M.C.); (I.P.); (S.C.)
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98166 Messina, Italy; (M.L.); (G.C.); (M.C.); (I.P.); (S.C.)
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98166 Messina, Italy; (M.L.); (G.C.); (M.C.); (I.P.); (S.C.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98166 Messina, Italy; (M.L.); (G.C.); (M.C.); (I.P.); (S.C.)
| | - Cristina Colarossi
- Mediterranean Institute of Oncology, Via Penninazzo 7, 95029 Viagrande, Italy; (C.C.); (M.M.)
| | - Marzia Mare
- Mediterranean Institute of Oncology, Via Penninazzo 7, 95029 Viagrande, Italy; (C.C.); (M.M.)
| | | | - Maria Caffo
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98166 Messina, Italy; (M.L.); (G.C.); (M.C.); (I.P.); (S.C.)
- Correspondence:
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98166 Messina, Italy; (M.L.); (G.C.); (M.C.); (I.P.); (S.C.)
| |
Collapse
|
155
|
Motohara T, Yoshida GJ, Katabuchi H. The hallmarks of ovarian cancer stem cells and niches: Exploring their harmonious interplay in therapy resistance. Semin Cancer Biol 2021; 77:182-193. [PMID: 33812986 DOI: 10.1016/j.semcancer.2021.03.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/20/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022]
Abstract
The concept of a "cancer stem cell" has evolved over the past decades, and research on cancer stem cell biology has entered into a stage of remarkable progress. Cancer stem cells are a major determining factor contributing to the establishment of phenotypic and functional intratumoral heterogeneity in synchronization with their surrounding "cancer stem cell niches." They serve as the driving force for cancer initiation, metastasis, and therapeutic resistance in various types of malignancies. In verity, reciprocal interplay between ovarian cancer stem cells and their niches involves a complex but ingeniously orchestrated tumor microenvironment within the intraperitoneal milieu and especially contribute to chemotherapy resistance in patients with advanced ovarian cancer. Herein, we review the principles of our current understanding of the biological features of ovarian cancer stem cells, focusing mainly on the precise mechanisms underlying acquired chemotherapy resistance. Furthermore, we highlight the specific roles of various cancer-associated stromal and immune cells in creating possible cancer stem cell niches that regulate ovarian cancer stemness.
Collapse
Affiliation(s)
- Takeshi Motohara
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto City, Kumamoto, 860-8556, Japan.
| | - Go J Yoshida
- Department of Immunological Diagnosis, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hidetaka Katabuchi
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto City, Kumamoto, 860-8556, Japan
| |
Collapse
|
156
|
Virtuoso A, Giovannoni R, De Luca C, Gargano F, Cerasuolo M, Maggio N, Lavitrano M, Papa M. The Glioblastoma Microenvironment: Morphology, Metabolism, and Molecular Signature of Glial Dynamics to Discover Metabolic Rewiring Sequence. Int J Mol Sci 2021; 22:3301. [PMID: 33804873 PMCID: PMC8036663 DOI: 10.3390/ijms22073301] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Different functional states determine glioblastoma (GBM) heterogeneity. Brain cancer cells coexist with the glial cells in a functional syncytium based on a continuous metabolic rewiring. However, standard glioma therapies do not account for the effects of the glial cells within the tumor microenvironment. This may be a possible reason for the lack of improvements in patients with high-grade gliomas therapies. Cell metabolism and bioenergetic fitness depend on the availability of nutrients and interactions in the microenvironment. It is strictly related to the cell location in the tumor mass, proximity to blood vessels, biochemical gradients, and tumor evolution, underlying the influence of the context and the timeline in anti-tumor therapeutic approaches. Besides the cancer metabolic strategies, here we review the modifications found in the GBM-associated glia, focusing on morphological, molecular, and metabolic features. We propose to analyze the GBM metabolic rewiring processes from a systems biology perspective. We aim at defining the crosstalk between GBM and the glial cells as modules. The complex networking may be expressed by metabolic modules corresponding to the GBM growth and spreading phases. Variation in the oxidative phosphorylation (OXPHOS) rate and regulation appears to be the most important part of the metabolic and functional heterogeneity, correlating with glycolysis and response to hypoxia. Integrated metabolic modules along with molecular and morphological features could allow the identification of key factors for controlling the GBM-stroma metabolism in multi-targeted, time-dependent therapies.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | | | - Ciro De Luca
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Francesca Gargano
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Michele Cerasuolo
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Nicola Maggio
- Department of Neurology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel;
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5211401, Israel
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Michele Papa
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
157
|
A DM1-doped porous gold nanoshell system for NIR accelerated redox-responsive release and triple modal imaging guided photothermal synergistic chemotherapy. J Nanobiotechnology 2021; 19:77. [PMID: 33741008 PMCID: PMC7976706 DOI: 10.1186/s12951-021-00824-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/06/2021] [Indexed: 12/14/2022] Open
Abstract
Background Although many treatments for breast cancer are available, poor tumour targeting limits the effectiveness of most approaches. Consequently, it is difficult to achieve satisfactory results with monotherapies. The lack of accurate diagnostic and monitoring methods also limit the benefits of cancer treatment. The aim of this study was to design a nanocarrier comprising porous gold nanoshells (PGNSs) co-decorated with methoxy polyethylene glycol (mPEG) and trastuzumab (Herceptin®, HER), a therapeutic monoclonal antibody that binds specifically to human epidermal receptor-2 (HER2)-overexpressing breast cancer cells. Furthermore, a derivative of the microtubule-targeting drug maytansine (DM1) was incorporated in the PGNSs. Methods Prepared PGNSs were coated with mPEG, DM1 and HER via electrostatic interactions and Au–S bonds to yield DM1-mPEG/HER-PGNSs. SK-BR-3 (high HER2 expression) and MCF-7 (low HER2) breast cancer cells were treated with DM1-mPEG/HER-PGNSs, and cytotoxicity was evaluated in terms of cell viability and apoptosis. The selective uptake of the coated PGNSs by cancer cells and subsequent intracellular accumulation were studied in vitro and in vivo using inductively coupled plasma mass spectrometry and fluorescence imaging. The multimodal imaging feasibility and synergistic chemo-photothermal therapeutic efficacy of the DM1-mPEG/HER-PGNSs were investigated in breast cancer tumour-bearing mice. The molecular mechanisms associated with the anti-tumour therapeutic use of the nanoparticles were also elucidated. Result The prepared DM1-mPEG/HER-PGNSs had a size of 78.6 nm and displayed excellent colloidal stability, photothermal conversion ability and redox-sensitive drug release. These DM1-mPEG/HER-PGNSs were taken up selectively by cancer cells in vitro and accumulated at tumour sites in vivo. Moreover, the DM1-mPEG/HER-PGNSs enhanced the performance of multimodal computed tomography (CT), photoacoustic (PA) and photothermal (PT) imaging and enabled chemo-thermal combination therapy. The therapeutic mechanism involved the induction of tumour cell apoptosis via the activation of tubulin, caspase-3 and the heat shock protein 70 pathway. M2 macrophage suppression and anti-metastatic functions were also observed. Conclusion The prepared DM1-mPEG/HER-PGNSs enabled nanodart-like tumour targeting, visibility by CT, PA and PT imaging in vivo and powerful tumour inhibition mediated by chemo-thermal combination therapy in vivo. In summary, these unique gold nanocarriers appear to have good potential as theranostic nanoagents that can serve both as a probe for enhanced multimodal imaging and as a novel targeted anti-tumour drug delivery system to achieve precision nanomedicine for cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00824-5.
Collapse
|
158
|
Biserova K, Jakovlevs A, Uljanovs R, Strumfa I. Cancer Stem Cells: Significance in Origin, Pathogenesis and Treatment of Glioblastoma. Cells 2021; 10:cells10030621. [PMID: 33799798 PMCID: PMC8000844 DOI: 10.3390/cells10030621] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/27/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs), known also as tumor-initiating cells, are quiescent, pluripotent, self-renewing neoplastic cells that were first identified in hematologic tumors and soon after in solid malignancies. CSCs have attracted remarkable research interest due to their role in tumor resistance to chemotherapy and radiation treatment as well as recurrence. Extensive research has been devoted to the role of CSCs in glioblastoma multiforme (GBM), the most common primary brain tumor in adults, which is characterized by a dismal prognosis because of its aggressive course and poor response to treatment. The aim of the current paper is to provide an overview of current knowledge on the role of cancer stem cells in the pathogenesis and treatment resistance of glioblastoma. The six regulatory mechanisms of glioma stem cells (GSCs)—tumor microenvironment, niche concept, metabolism, immunity, genetics, and epigenetics—are reviewed. The molecular markers used to identify GSCs are described. The role of GSCs in the treatment resistance of glioblastoma is reviewed, along with future treatment options targeting GSCs. Stem cells of glioblastoma thus represent both a driving mechanism of major treatment difficulties and a possible target for more effective future approaches.
Collapse
Affiliation(s)
- Karina Biserova
- Faculty of Residency, Riga Stradins University, 16 Dzirciema Street, LV-1007 Riga, Latvia
- Correspondence:
| | - Arvids Jakovlevs
- Department of Pathology, Riga Stradins University, 16 Dzirciema Street, LV-1007 Riga, Latvia; (A.J.); (R.U.); (I.S.)
| | - Romans Uljanovs
- Department of Pathology, Riga Stradins University, 16 Dzirciema Street, LV-1007 Riga, Latvia; (A.J.); (R.U.); (I.S.)
| | - Ilze Strumfa
- Department of Pathology, Riga Stradins University, 16 Dzirciema Street, LV-1007 Riga, Latvia; (A.J.); (R.U.); (I.S.)
| |
Collapse
|
159
|
Yeo ECF, Brown MP, Gargett T, Ebert LM. The Role of Cytokines and Chemokines in Shaping the Immune Microenvironment of Glioblastoma: Implications for Immunotherapy. Cells 2021; 10:607. [PMID: 33803414 PMCID: PMC8001644 DOI: 10.3390/cells10030607] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/23/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is the most common form of primary brain tumour in adults. For more than a decade, conventional treatment has produced a relatively modest improvement in the overall survival of glioblastoma patients. The immunosuppressive mechanisms employed by neoplastic and non-neoplastic cells within the tumour can limit treatment efficacy, and this can include the secretion of immunosuppressive cytokines and chemokines. These factors can play a significant role in immune modulation, thus disabling anti-tumour responses and contributing to tumour progression. Here, we review the complex interplay between populations of immune and tumour cells together with defined contributions by key cytokines and chemokines to these intercellular interactions. Understanding how these tumour-derived factors facilitate the crosstalk between cells may identify molecular candidates for potential immunotherapeutic targeting, which may enable better tumour control and improved patient survival.
Collapse
Affiliation(s)
- Erica C. F. Yeo
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia; (E.C.F.Y.); (M.P.B.); (T.G.)
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Michael P. Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia; (E.C.F.Y.); (M.P.B.); (T.G.)
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Tessa Gargett
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia; (E.C.F.Y.); (M.P.B.); (T.G.)
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Lisa M. Ebert
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia; (E.C.F.Y.); (M.P.B.); (T.G.)
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
160
|
Wang Q, Zhang J, Fang S, Wang J, Han X, Liu F, Jin G. P4HA1 Down-Regulation Inhibits Glioma Invasiveness by Promoting M1 Microglia Polarization. Onco Targets Ther 2021; 14:1771-1782. [PMID: 33727827 PMCID: PMC7954035 DOI: 10.2147/ott.s299977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/16/2021] [Indexed: 12/09/2022] Open
Abstract
Background Polarization of microglia cells in the glioma microenvironment is closely related to the malignant progression and invasion of gliomas. Prolyl 4-hydroxylase subunit α1 (P4HA1) is the rate-limiting subunit of prolyl 4-hydroxylase (P4H). In previous studies, we showed that P4HA1 could promote the proliferation, migration, and invasion of glioma cells, but the specific mechanisms through which this occurs have not been fully elucidated. Materials and Methods Interactions between glioma and microglia cells were analyzed using bioinformatics. Then, co-culture models were used to obtain conditioned media. To characterize microglial cell polarization, we used PCR and immunofluorescence. Proliferation and invasion assays were used to explore the biological behavior of glioma cells affected by microglia. Finally, marker expression was detected using immunohistochemistry in glioblastoma multiform (GBM) specimens. Results Knockdown of P4HA1 resulted in reduced chemotaxis of microglia toward GBM cells and increased polarization of microglia toward the M1 phenotype. The changed microglial polarization state, in turn, inhibited the proliferation and invasion of GBM cells. Moreover, in GBM tissue specimens, the P4HA1 expression level is negatively correlated with that of the CD86 microglia M1-specific marker. Conclusion Our results show that P4HA1 promotes immunosuppressive microenvironment formation by cross-talk between GBM and microglia cells and indirectly increases the aggressiveness of GBM.
Collapse
Affiliation(s)
- Qiyan Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| | - Xiangming Han
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 10070, People's Republic of China
| |
Collapse
|
161
|
Lorimer IAJ. Aberrant Rac pathway signalling in glioblastoma. Small GTPases 2021; 12:81-95. [PMID: 31032735 PMCID: PMC7849730 DOI: 10.1080/21541248.2019.1612694] [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: 03/11/2019] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022] Open
Abstract
Glioblastoma is an aggressive and incurable form of brain cancer. Both mutation analysis in human glioblastoma and mouse modelling studies have shown that aberrant activation of the PI 3-kinase pathway is a central driver of glioblastoma malignancy. The small GTPase Rac is activated downstream of this pathway, mediating a subset of the effects of aberrant PI 3-kinase pathway activation. Here I discuss the current state of our knowledge on Rac activation mechanisms in glioblastoma. Current knowledge on roles for specific PI 3-kinase pathway responsive Rac guanine nucleotide exchange factors in glioblastoma is reviewed. Rac is best known for its role in promoting cell motility and invasion, but there is also evidence for roles in multiple other cellular processes with cancer relevance, including proliferation, differentiation, apoptosis, DNA damage responses, metabolism, angiogenesis and immunosuppression. I review what is known about the role of Rac in these processes in glioblastoma. Finally, I assess possible strategies to inhibit this pathway in glioblastoma through either direct inhibition of Rac or inhibition of upstream activators or downstream mediators of Rac signalling.
Collapse
Affiliation(s)
- Ian AJ Lorimer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
162
|
Gao X, Li S, Ding F, Liu X, Wu Y, Li J, Feng J, Zhu X, Zhang C. A Virus-Mimicking Nucleic Acid Nanogel Reprograms Microglia and Macrophages for Glioblastoma Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006116. [PMID: 33501743 DOI: 10.1002/adma.202006116] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/19/2020] [Indexed: 06/12/2023]
Abstract
Immunotherapy is recognized as one of the most promising approaches to treat cancers. However, its effect in glioblastoma (GBM) treatment is insufficient, which can in part be attributed to the immunosuppressive tumor microenvironment (TME). Microglia and macrophages are the main immune infiltrating cells in the TME of GBM. Unfortunately, instead of initiating the anti-tumor response, GBM-infiltrating microglia and macrophages switch to a tumor-promoting phenotype (M2), and support tumor growth, angiogenesis, and immunosuppression by the release of cytokines. In this work, a virus-mimicking membrane-coated nucleic acid nanogel Vir-Gel embedded with therapeutic miRNA is developed, which can reprogram microglia and macrophages from a pro-invasive M2 phenotype to an anti-tumor M1 phenotype. By mimicking the virus infection process, Vir-Gel significantly enhances the targetability and cell uptake efficiency of the miR155-bearing nucleic acid nanogel. In vivo evaluations demonstrate that Vir-Gel apparently prolongs the circulation lifetime of miR155 and endows it with an active tumor-targeting capability and excellent tumor inhibition efficacy. Owing to its noninvasive feature and effective delivery capability, the virus-mimicking nucleic acid nanogel provides a general and convenient platform that can successfully treat a wide range of diseases.
Collapse
Affiliation(s)
- Xihui Gao
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200 240, China
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, 6600th Nanfeng Road, Fengxian District, Shanghai, 201 499, China
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200 032, China
| | - Sha Li
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, 6600th Nanfeng Road, Fengxian District, Shanghai, 201 499, China
| | - Fei Ding
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200 240, China
| | - Xinlong Liu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200 240, China
| | - Yijing Wu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200 240, China
| | - Jing Li
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, 6600th Nanfeng Road, Fengxian District, Shanghai, 201 499, China
| | - Jing Feng
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, 6600th Nanfeng Road, Fengxian District, Shanghai, 201 499, China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200 240, China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200 240, China
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, 6600th Nanfeng Road, Fengxian District, Shanghai, 201 499, China
| |
Collapse
|
163
|
Hasanpour Segherlou Z, Nouri-Vaskeh M, Noroozi Guilandehi S, Baghbanzadeh A, Zand R, Baradaran B, Zarei M. GDF-15: Diagnostic, prognostic, and therapeutic significance in glioblastoma multiforme. J Cell Physiol 2021; 236:5564-5581. [PMID: 33580506 DOI: 10.1002/jcp.30289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the commonest primary malignant brain tumor and has a remarkably weak prognosis. According to the aggressive form of GBM, understanding the accurate molecular mechanism associated with GBM pathogenesis is essential. Growth differentiation factor 15 (GDF-15) belongs to transforming growth factor-β superfamily with important roles to control biological processes. It affects cancer growth and progression, drug resistance, and metastasis. It also can promote stemness in many cancers, and also can stress reactions control, bone generation, hematopoietic growth, adipose tissue performance, and body growth, and contributes to cardiovascular disorders. The role GDF-15 to develop and progress cancer is complicated and remains unclear. GDF-15 possesses tumor suppressor properties, as well as an oncogenic effect. GDF-15 antitumorigenic and protumorigenic impacts on tumor development are linked to the cancer type and stage. However, the GDF-15 signaling and mechanism have not yet been completely identified because of no recognized cognate receptor.
Collapse
Affiliation(s)
| | - Masoud Nouri-Vaskeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Zand
- Department of Neurology, Geisinger Health System, Danville, Pennsylvania, USA
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Zarei
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
164
|
Wei J, Chen P, Gupta P, Ott M, Zamler D, Kassab C, Bhat KP, Curran MA, de Groot JF, Heimberger AB. Immune biology of glioma-associated macrophages and microglia: functional and therapeutic implications. Neuro Oncol 2021; 22:180-194. [PMID: 31679017 DOI: 10.1093/neuonc/noz212] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
CNS immune defenses are marshaled and dominated by brain resident macrophages and microglia, which are the innate immune sentinels and frontline host immune barriers against various pathogenic insults. These myeloid lineage cells are the predominant immune population in gliomas and can constitute up to 30-50% of the total cellular composition. Parenchymal microglial cells and recruited monocyte-derived macrophages from the periphery exhibit disease-specific phenotypic characteristics with spatial and temporal distinctions and are heterogeneous subpopulations based on their molecular signatures. A preponderance of myeloid over lymphoid lineage cells during CNS inflammation, including gliomas, is a contrasting feature of brain immunity relative to peripheral immunity. Herein we discuss glioma-associated macrophage and microglia immune biology in the context of their identity, molecular drivers of recruitment, nomenclature and functional paradoxes, therapeutic reprogramming and polarization strategies, relevant challenges, and our perspectives on therapeutic modulation.
Collapse
Affiliation(s)
- Jun Wei
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peiwen Chen
- Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pravesh Gupta
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Martina Ott
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Zamler
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia Kassab
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krishna P Bhat
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Curran
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John F de Groot
- Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
165
|
Zhang Y, Xie Y, He L, Tang J, He Q, Cao Q, Cui L, Guo W, Hua K, Dimberg A, Wang L, Zhang L. 1p/19q co-deletion status is associated with distinct tumor-associated macrophage infiltration in IDH mutated lower-grade gliomas. Cell Oncol (Dordr) 2021; 44:193-204. [PMID: 32915415 DOI: 10.1007/s13402-020-00561-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAM)s are critical regulators of glioma progression. As yet, however, TAMs in isocitrate dehydrogenase (IDH) mutated lower-grade gliomas (LGGs) have not been thoroughly investigated. The aim of this study was to determine whether 1p/19q co-deletion status affects the TAM phenotype or its prevalence in IDH mutated LGGs. METHODS TAMs in IDH mutated LGGs were analyzed using transcriptome data from 230 samples in the TCGA database in combination with transcriptome data from single-cell RNA sequencing of IDH-mutated LGGs. Proteins potentially involved in TAM regulation were examined by immuno-staining in primary LGG samples harboring IDH mutations. Essential signaling pathways regulating TAM phenotypes were investigated in a glioma mouse model using small molecule inhibitors. RESULTS Most of the TAMs in IDH-mutated LGGs expressed the M1 activation markers CD86 and TNF, whereas a subset of individual TAMs co-expressed both M1 and M2-related markers. Bioinformatics analysis in combination with immuno-staining of IDH-mutated patient samples revealed higher amounts of TAMs expressing M2-related markers in 1p/19q non-codeletion IDH-mutated LGGs compared to 1p/19q codeletion LGGs. The levels of transforming growth factor beta 1 (TGFβ1) and macrophage colony-stimulating factor (M-CSF) were significantly higher in 1p/19q non-codeletion LGGs than in 1p/19q codeletion LGGs. M-CSF and TGFβ1 signal inhibition decreased tumor growth and modulated the TAM phenotype in a glioma mouse model. CONCLUSIONS Our data indicate that 1p/19q co-deletion status relates to distinct TAM infiltration in gliomas, which is likely mediated by M-CSF and TGFβ1 signaling. M-CSF and TGFβ1 signaling may play a pivotal role in regulating the TAM phenotype in glioma.
Collapse
Affiliation(s)
- Yanyu Zhang
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Box 582, BMC, Husargatan 3, 75123, Uppsala, Sweden
| | - Yuan Xie
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Liqun He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuro-injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Jiefu Tang
- Department of Spine Surgery, Huaihua No.2 Hospital, Hunan University of Medicine, Huaihua, 418000, China
| | - Qiyuan He
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Qingze Cao
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Langjun Cui
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University of PLA (the Fourth Military Medical University), 569 Xinsi Road, Xi'an, 710038, China
| | - Kai Hua
- The College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University of PLA (the Fourth Military Medical University), 569 Xinsi Road, Xi'an, 710038, China.
| | - Lei Zhang
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China.
- Department of Spine Surgery, Huaihua No.2 Hospital, Hunan University of Medicine, Huaihua, 418000, China.
| |
Collapse
|
166
|
Ou A, Ott M, Fang D, Heimberger AB. The Role and Therapeutic Targeting of JAK/STAT Signaling in Glioblastoma. Cancers (Basel) 2021; 13:437. [PMID: 33498872 PMCID: PMC7865703 DOI: 10.3390/cancers13030437] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma remains one of the deadliest and treatment-refractory human malignancies in large part due to its diffusely infiltrative nature, molecular heterogeneity, and capacity for immune escape. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway contributes substantively to a wide variety of protumorigenic functions, including proliferation, anti-apoptosis, angiogenesis, stem cell maintenance, and immune suppression. We review the current state of knowledge regarding the biological role of JAK/STAT signaling in glioblastoma, therapeutic strategies, and future directions for the field.
Collapse
Affiliation(s)
- Alexander Ou
- Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA;
| | - Martina Ott
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (M.O.); (D.F.)
| | - Dexing Fang
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (M.O.); (D.F.)
| | - Amy B. Heimberger
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (M.O.); (D.F.)
| |
Collapse
|
167
|
Chen P, Hsu WH, Han J, Xia Y, DePinho RA. Cancer Stemness Meets Immunity: From Mechanism to Therapy. Cell Rep 2021; 34:108597. [PMID: 33406434 PMCID: PMC7839836 DOI: 10.1016/j.celrep.2020.108597] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/24/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs) are self-renewing cells that facilitate tumor initiation, promote metastasis, and enhance cancer therapy resistance. Transcriptomic analyses across many cancer types have revealed a prominent association between stemness and immune signatures, potentially implying a biological interaction between such hallmark features of cancer. Emerging experimental evidence has substantiated the influence of CSCs on immune cells, including tumor-associated macrophages, myeloid-derived suppressor cells, and T cells, in the tumor microenvironment and, reciprocally, the importance of such immune cells in sustaining CSC stemness and its survival niche. This review covers the cellular and molecular mechanisms underlying the symbiotic interactions between CSCs and immune cells and how such heterotypic signaling maintains a tumor-promoting ecosystem and informs therapeutic strategies intercepting this co-dependency.
Collapse
Affiliation(s)
- Peiwen Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jincheng Han
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Xia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
168
|
Toedebusch R, Grodzki AC, Dickinson PJ, Woolard K, Vinson N, Sturges B, Snyder J, Li CF, Nagasaka O, Consales B, Vernau K, Knipe M, Murthy V, Lein PJ, Toedebusch CM. Glioma-associated microglia/macrophages augment tumorigenicity in canine astrocytoma, a naturally occurring model of human glioma. Neurooncol Adv 2021; 3:vdab062. [PMID: 34131649 PMCID: PMC8193901 DOI: 10.1093/noajnl/vdab062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Glioma-associated microglia/macrophages (GAMs) markedly influence glioma progression. Under the influence of transforming growth factor beta (TGFB), GAMs are polarized toward a tumor-supportive phenotype. However, neither therapeutic targeting of GAM recruitment nor TGFB signaling demonstrated efficacy in glioma patients despite efficacy in preclinical models, underscoring the need for a comprehensive understanding of the TGFB/GAM axis. Spontaneously occurring canine gliomas share many features with human glioma and provide a complementary translational animal model for further study. Given the importance of GAM and TGFB in human glioma, the aims of this study were to further define the GAM-associated molecular profile and the relevance of TGFB signaling in canine glioma that may serve as the basis for future translational studies. METHODS GAM morphometry, levels of GAM-associated molecules, and the canonical TGFB signaling axis were compared in archived samples of canine astrocytomas versus normal canine brain. Furthermore, the effect of TGFB on the malignant phenotype of canine astrocytoma cells was evaluated. RESULTS GAMs diffusely infiltrated canine astrocytomas. GAM density was increased in high-grade tumors that correlated with a pro-tumorigenic molecular signature and upregulation of the canonical TGFB signaling axis. Moreover, TGFB1 enhanced the migration of canine astrocytoma cells in vitro. CONCLUSIONS Canine astrocytomas share a similar GAM-associated immune landscape with human adult glioma. Our data also support a contributing role for TGFB1 signaling in the malignant phenotype of canine astrocytoma. These data further support naturally occurring canine glioma as a valid model for the investigation of GAM-associated therapeutic strategies for human malignant glioma.
Collapse
Affiliation(s)
- Ryan Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Ana Cristina Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Peter J Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Kevin Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Nicole Vinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Beverly Sturges
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - John Snyder
- Riemann Computing, LLC, St. Louis, Missouri, USA
| | - Chai-Fei Li
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Ori Nagasaka
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Blaire Consales
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Karen Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Marguerite Knipe
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Vishal Murthy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Christine M Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
169
|
Stephen ZR, Zhang M. Recent Progress in the Synergistic Combination of Nanoparticle-Mediated Hyperthermia and Immunotherapy for Treatment of Cancer. Adv Healthc Mater 2021; 10:e2001415. [PMID: 33236511 PMCID: PMC8034553 DOI: 10.1002/adhm.202001415] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has demonstrated great clinical success in certain cancers, driven primarily by immune checkpoint blockade and adoptive cell therapies. Immunotherapy can elicit strong, durable responses in some patients, but others do not respond, and to date immunotherapy has demonstrated success in only a limited number of cancers. To address this limitation, combinatorial approaches with chemo- and radiotherapy have been applied in the clinic. Extensive preclinical evidence suggests that hyperthermia therapy (HT) has considerable potential to augment immunotherapy with minimal toxicity. This progress report will provide a brief overview of immunotherapy and HT approaches and highlight recent progress in the application of nanoparticle (NP)-based HT in combination with immunotherapy. NPs allow for tumor-specific targeting of deep tissue tumors while potentially providing more even heating. NP-based HT increases tumor immunogenicity and tumor permeability, which improves immune cell infiltration and creates an environment more responsive to immunotherapy, particularly in solid tumors.
Collapse
Affiliation(s)
- Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, Department of Neurological Surgery, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
170
|
Boyd NH, Tran AN, Bernstock JD, Etminan T, Jones AB, Gillespie GY, Friedman GK, Hjelmeland AB. Glioma stem cells and their roles within the hypoxic tumor microenvironment. Theranostics 2021; 11:665-683. [PMID: 33391498 PMCID: PMC7738846 DOI: 10.7150/thno.41692] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor microenvironments are the result of cellular alterations in cancer that support unrestricted growth and proliferation and result in further modifications in cell behavior, which are critical for tumor progression. Angiogenesis and therapeutic resistance are known to be modulated by hypoxia and other tumor microenvironments, such as acidic stress, both of which are core features of the glioblastoma microenvironment. Hypoxia has also been shown to promote a stem-like state in both non-neoplastic and tumor cells. In glial tumors, glioma stem cells (GSCs) are central in tumor growth, angiogenesis, and therapeutic resistance, and further investigation of the interplay between tumor microenvironments and GSCs is critical to the search for better treatment options for glioblastoma. Accordingly, we summarize the impact of hypoxia and acidic stress on GSC signaling and biologic phenotypes, and potential methods to inhibit these pathways.
Collapse
|
171
|
Ou A, Yung WKA, Majd N. Molecular Mechanisms of Treatment Resistance in Glioblastoma. Int J Mol Sci 2020; 22:E351. [PMID: 33396284 PMCID: PMC7794986 DOI: 10.3390/ijms22010351] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults and is almost invariably fatal. Despite our growing understanding of the various mechanisms underlying treatment failure, the standard-of-care therapy has not changed over the last two decades, signifying a great unmet need. The challenges of treating glioblastoma are many and include inadequate drug or agent delivery across the blood-brain barrier, abundant intra- and intertumoral heterogeneity, redundant signaling pathways, and an immunosuppressive microenvironment. Here, we review the innate and adaptive molecular mechanisms underlying glioblastoma's treatment resistance, emphasizing the intrinsic challenges therapeutic interventions must overcome-namely, the blood-brain barrier, tumoral heterogeneity, and microenvironment-and the mechanisms of resistance to conventional treatments, targeted therapy, and immunotherapy.
Collapse
Affiliation(s)
| | - W. K. Alfred Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 431, Houston, TX 77030, USA;
| | - Nazanin Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 431, Houston, TX 77030, USA;
| |
Collapse
|
172
|
Abstract
Tumor progression is profoundly influenced by interactions between cancer cells and the tumor microenvironment (TME). Among the various non-neoplastic cells present, immune cells are critical players in tumor development and have thus emerged as attractive therapeutic targets. Malignant gliomas exhibit a unique immune landscape characterized by high numbers of tumor-associated macrophages (TAMs). Despite encouraging preclinical results, targeting TAMs has yielded limited clinical success as a strategy for slowing glioma progression. The slow translational progress of TAM-targeted therapies is due in part to an incomplete understanding of the factors driving TAM recruitment, differentiation, and polarization. Furthermore, the functions that TAMs adopt in gliomas remain largely unknown. Progress in addressing these gaps requires sophisticated culture platforms capable of capturing key cellular and physical TME features. This review summarizes the current understanding of TAMs in gliomas and highlights the utility of in vitro TME models for investigating TAM-cancer cell cross talk.
Collapse
Affiliation(s)
- Erin A. Akins
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Manish K. Aghi
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sanjay Kumar
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
173
|
Marisetty A, Wei J, Kong LY, Ott M, Fang D, Sabbagh A, Heimberger AB. MiR-181 Family Modulates Osteopontin in Glioblastoma Multiforme. Cancers (Basel) 2020; 12:cancers12123813. [PMID: 33348707 PMCID: PMC7765845 DOI: 10.3390/cancers12123813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary MicroRNAs can silence a broad set of target genes that may benefit heterogeneous tumors like glioblastoma. We have previously shown that osteopontin has an oncogenic role and may have immune modulatory effects on macrophages. In the current study, we used miRNAs to target osteopontin in tumor cells and modulate immune cells to elicit an antitumor effect. Intravenous delivery of miR-181a to immune competent mice bearing intracranial glioblastoma demonstrated a 22% increase in median survival duration relative to that of control mice. The overexpression of miR-181a in tumor cells led to decreased OPN production and proliferation and increased apoptosis in vitro, and increased survival duration of the mice when compared to its controls. miR-181a controls osteopontin expression in tumor cells by regulating their proliferation and apoptosis. Abstract MiRNAs can silence a wide range of genes, which may be an advantage for targeting heterogenous tumors like glioblastoma. Osteopontin (OPN) plays both an oncogenic role in a variety of cancers and can immune modulate macrophages. We conducted a genome wide profiling and bioinformatic analysis to identify miR-181a/b/c/d as potential miRNAs that target OPN. Luciferase assays confirmed the binding potential of miRNAs to OPN. Expression levels of miR-181a/b/c/d and OPN were evaluated by using quantitative real-time PCR and enzyme-linked immunosorbent assay in mouse and human glioblastomas and macrophages that showed these miRNAs were downregulated in Glioblastoma associated CD11b+ cells compared to their matched blood CD14b+ cells. miRNA mimicking and overexpression using lentiviruses showed that MiR-181a overexpression in glioblastoma cells led to decreased OPN production and proliferation and increased apoptosis in vitro. MiR-181a treatment of immune competent mice bearing intracranial glioblastoma demonstrated a 22% increase in median survival duration relative to that of control mice.
Collapse
|
174
|
Timmerman R, Burm SM, Bajramovic JJ. Tissue-specific features of microglial innate immune responses. Neurochem Int 2020; 142:104924. [PMID: 33248205 DOI: 10.1016/j.neuint.2020.104924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
As tissue-resident macrophages of the brain, microglia are increasingly considered as cellular targets for therapeutical intervention. Innate immune responses in particular have been implicated in central nervous system (CNS) infections, neuro-oncology, neuroinflammatory and neurodegenerative diseases. We here review the impact of 'nature and nurture' on microglial innate immune responses and summarize documented tissue-specific adaptations. Overall, such adaptations are associated with regulatory processes rather than with overt differences in the expressed repertoire of activating receptors of different tissue-resident macrophages. Microglial responses are characterized by slower kinetics, by a more persistent nature and by a differential usage of downstream enzymes and accessory receptors. We further consider factors like aging, previous exposure to inflammatory stimuli, and differences in the microenvironment that can modulate innate immune responses. The long-life span of microglia in the metabolically active CNS renders them susceptible to the phenomenon of 'inflammaging', and major challenges lie in the unraveling of the factors that underlie age-related alterations in microglial behavior.
Collapse
Affiliation(s)
- R Timmerman
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - S M Burm
- Genmab, Utrecht, the Netherlands
| | - J J Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands.
| |
Collapse
|
175
|
Salminen A. Hypoperfusion is a potential inducer of immunosuppressive network in Alzheimer's disease. Neurochem Int 2020; 142:104919. [PMID: 33242538 DOI: 10.1016/j.neuint.2020.104919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/12/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease which causes a non-reversible cognitive impairment and dementia. The primary cause of late-onset AD remains unknown although its pathology was discovered over a century ago. Recently, the vascular hypothesis of AD has received backing from evidence emerging from neuroimaging studies which have revealed the presence of a significant hypoperfusion in the brain regions vulnerable to AD pathology. In fact, hypoxia can explain many of the pathological changes evident in AD pathology, e.g. the deposition of β-amyloid plaques and chronic low-grade inflammation. Hypoxia-inducible factor-1α (HIF-1α) stimulates inflammatory responses and modulates both innate and adaptive immunity. It is known that hypoxia-induced inflammation evokes compensatory anti-inflammatory response involving tissue-resident microglia/macrophages and infiltrated immune cells. Hypoxia/HIF-1α induce immunosuppression by (i) increasing the expression of immunosuppressive genes, (ii) stimulating adenosinergic signaling, (iii) enhancing aerobic glycolysis, i.e. lactate production, and (iv) augmenting the secretion of immunosuppressive exosomes. Interestingly, it seems that these common mechanisms are also involved in the pathogenesis of AD. In AD pathology, an enhanced immunosuppression appears, e.g. as a shift in microglia/macrophage phenotypes towards the anti-inflammatory M2 phenotype and an increase in the numbers of regulatory T cells (Treg). The augmented anti-inflammatory capacity promotes the resolution of acute inflammation but persistent inflammation has crucial effects not only on immune cells but also harmful responses to the homeostasis of AD brain. I will examine in detail the mechanisms of the hypoperfusion/hypoxia-induced immunosuppressive state in general and especially, in its association with AD pathogenesis. These immunological observations support the vascular hypothesis of AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
176
|
Zhang Q, Han Z, Zhu Y, Chen J, Li W. Role of hypoxia inducible factor-1 in cancer stem cells (Review). Mol Med Rep 2020; 23:17. [PMID: 33179080 PMCID: PMC7673349 DOI: 10.3892/mmr.2020.11655] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) have been found to play a decisive role in cancer recurrence, metastasis, and chemo‑, radio‑ and immuno‑resistance. Understanding the mechanism of CSC self‑renewal and proliferation may help overcome the limitations of clinical treatment. The microenvironment of tumor growth consists of a lack of oxygen, and hypoxia has been confirmed to induce cancer cell invasion, metastasis and epithelial‑mesenchymal transition, and is usually associated with poor prognosis and low survival rates. Hypoxia inducible factor‑1 (HIF‑1) can be stably expressed under hypoxia and act as an important molecule to regulate the development of CSCs, but the specific mechanism remains unclear. The present review attempted to explain the role of HIF‑1 in the generation and maintenance of CSCs from the perspective of epigenetics, metabolic reprogramming, tumor immunity, CSC markers, non‑coding RNA and signaling pathways associated with HIF‑1, in order to provide novel targets with HIF‑1 as the core for clinical treatment, and extend the life of patients.
Collapse
Affiliation(s)
- Qi Zhang
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Zhenzhen Han
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Yanbo Zhu
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Jingcheng Chen
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Wei Li
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| |
Collapse
|
177
|
Radin DP, Tsirka SE. Interactions between Tumor Cells, Neurons, and Microglia in the Glioma Microenvironment. Int J Mol Sci 2020; 21:E8476. [PMID: 33187183 PMCID: PMC7698134 DOI: 10.3390/ijms21228476] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Despite significant strides made in understanding the pathophysiology of high-grade gliomas over the past two decades, most patients succumb to these neoplasias within two years of diagnosis. Furthermore, there are various co-morbidities associated with glioma and standard of care treatments. Emerging evidence suggests that aberrant glutamate secretion in the glioma microenvironment promotes tumor progression and contributes to the development of co-morbidities, such as cognitive defects, epilepsy, and widespread neurodegeneration. Recent data clearly illustrate that neurons directly synapse onto glioma cells and drive their proliferation and spread via glutamatergic action. Microglia are central nervous system-resident myeloid cells, modulate glioma growth, and possess the capacity to prune synapses and encourage synapse formation. However, current literature has yet to investigate the potential role of microglia in shaping synapse formation between neurons and glioma cells. Herein, we present the literature concerning glutamate's role in glioma progression, involving hyperexcitability and excitotoxic cell death of peritumoral neurons and stimulation of glioma proliferation and invasion. Furthermore, we discuss instances in which microglia are more likely to sculpt or encourage synapse formation during glioma treatment and propose studies to delineate the role of microglia in synapse formation between neurons and glioma cells. The sex-dependent oncogenic or oncolytic actions of microglia and myeloid cells, in general, are considered in addition to the functional differences between microglia and macrophages in tumor progression. We also put forth tractable methods to safely perturb aberrant glutamatergic action in the tumor microenvironment without significantly increasing the toxicities of the standard of care therapies for glioma therapy.
Collapse
Affiliation(s)
| | - Stella E. Tsirka
- Stony Brook Medical Scientist Training Program, Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, NY 11794-8651, USA;
| |
Collapse
|
178
|
Explicating the Pivotal Pathogenic, Diagnostic, and Therapeutic Biomarker Potentials of Myeloid-Derived Suppressor Cells in Glioblastoma. DISEASE MARKERS 2020; 2020:8844313. [PMID: 33204365 PMCID: PMC7657691 DOI: 10.1155/2020/8844313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/19/2020] [Accepted: 10/24/2020] [Indexed: 11/17/2022]
Abstract
Glioblastoma (GBM) is a malignant and aggressive central nervous tumor that originates from astrocytes. These pathogenic astrocytes divide rapidly and are sustained by enormous network of blood vessels via which they receive requisite nutrients. It well proven that GBM microenvironment is extremely infiltrated by myeloid-derived suppressor cells (MDSCs). MDSCs are a heterogeneous cluster of immature myeloid progenitors. They are key mediates in immune suppression as well as sustenance glioma growth, invasion, vascularization, and upsurge of regulatory T cells via different molecules. MDSCs are often elevated in the peripheral blood of patients with GBM. MDSCs in the peripheral blood as well as those infiltrating the GBM microenvironment correlated with poor prognosis. Also, an upsurge in circulating MDSCs in the peripheral blood of patients with GBM was observed compared to benign and grade I/II glioma patients. GBM patients with good prognosis presented with reduced MDSCs as well as augmented dendritic cells. Almost all chemotherapeutic medication for GBM has shown no obvious improvement in overall survival in patients. Nevertheless, low-dose chemotherapies were capable of suppressing the levels of MDSCs in GBM as well as multiple tumor models with metastatic to the brain. Thus, MDSCs are potential diagnostic as well as therapeutic biomarkers for GBM patients.
Collapse
|
179
|
Bajetto A, Thellung S, Dellacasagrande I, Pagano A, Barbieri F, Florio T. Cross talk between mesenchymal and glioblastoma stem cells: Communication beyond controversies. Stem Cells Transl Med 2020; 9:1310-1330. [PMID: 32543030 PMCID: PMC7581451 DOI: 10.1002/sctm.20-0161] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from bone marrow or other adult tissues (adipose tissue, dental pulp, amniotic fluid, and umbilical cord). In vitro, MSCs grow as adherent cells, display fibroblast-like morphology, and self-renew, undergoing specific mesodermal differentiation. High heterogeneity of MSCs from different origin, and differences in preparation techniques, make difficult to uniform their functional properties for therapeutic purposes. Immunomodulatory, migratory, and differentiation ability, fueled clinical MSC application in regenerative medicine, whereas beneficial effects are currently mainly ascribed to their secretome and extracellular vesicles. MSC translational potential in cancer therapy exploits putative anti-tumor activity and inherent tropism toward tumor sites to deliver cytotoxic drugs. However, controversial results emerged evaluating either the therapeutic potential or homing efficiency of MSCs, as both antitumor and protumor effects were reported. Glioblastoma (GBM) is the most malignant brain tumor and its development and aggressive nature is sustained by cancer stem cells (CSCs) and the identification of effective therapeutic is required. MSC dualistic action, tumor-promoting or tumor-targeting, is dependent on secreted factors and extracellular vesicles driving a complex cross talk between MSCs and GBM CSCs. Tumor-tropic ability of MSCs, besides providing an alternative therapeutic approach, could represent a tool to understand the biology of GBM CSCs and related paracrine mechanisms, underpinning MSC-GBM interactions. In this review, recent findings on the complex nature of MSCs will be highlighted, focusing on their elusive impact on GBM progression and aggressiveness by direct cell-cell interaction and via secretome, also facing the perspectives and challenges in treatment strategies.
Collapse
Affiliation(s)
- Adriana Bajetto
- Dipartimento di Medicina InternaUniversità di GenovaGenovaItaly
| | | | | | - Aldo Pagano
- Dipartimento di Medicina SperimentaleUniversità di GenovaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
| | | | - Tullio Florio
- Dipartimento di Medicina InternaUniversità di GenovaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
180
|
Efficacy of a novel double-controlled oncolytic adenovirus driven by the Ki67 core promoter and armed with IL-15 against glioblastoma cells. Cell Biosci 2020; 10:124. [PMID: 33133514 PMCID: PMC7592588 DOI: 10.1186/s13578-020-00485-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Background Glioblastoma (GBM) is an immunosuppressive, highly vascular and devastating malignant brain tumor. Even with progressive combination treatment that includes surgery, radiotherapy, and chemotherapy, the prognosis for GBM patients is still extremely poor. Oncolytic adenovirus (OAd) can specifically replicate in GBM cells, permitting the rapid copy of the therapeutic genes it carries. Moreover, E1A is an essential gene in adenoviral replication and is the first gene expressed upon viral infection. E1A expression can be regulated by the Ki67 promoter, while the CMV promoter drives therapeutic gene expression. However, the efficacy of a double-controlled OAd driven by the Ki67 core promoter and armed with IL-15 against GBM cells has not been investigated. Methods Fluorescence microscopy was performed to evaluate infection ability in the viruses. Cell viability was detected by CCK-8 assay. Levels of cytokines in different supernatants were determined by ELISA, and IL-15 gene expression was measured by RT-PCR. Angiogenic capacity was analyzed by tube formation assay. Results We successfully constructed a double-controlled oncolytic adenovirus driven by the Ki67 core promoter and armed with IL-15 that selectively infected and killed GBM cells while sparing normal cells. The adenoviruses prime IL-15 gene expression to significantly enhance anti-GBM efficacy through effective activation of microglial cells. Moreover, OAd not only directly inhibits angiogenesis but exhibits potent antiangiogenic capacity mediated by the reduction of VEGF secretion. Conclusions These results provide new insight into the effects of a novel double-controlled OAd driven by the Ki67 core promoter and armed with IL-15 in glioblastoma treatment, which may help in the development of novel therapies in solid tumors.
Collapse
|
181
|
Bier A, Hong X, Cazacu S, Goldstein H, Rand D, Xiang C, Jiang W, Ben-Asher HW, Attia M, Brodie A, She R, Poisson LM, Brodie C. miR-504 modulates the stemness and mesenchymal transition of glioma stem cells and their interaction with microglia via delivery by extracellular vesicles. Cell Death Dis 2020; 11:899. [PMID: 33093452 PMCID: PMC7581800 DOI: 10.1038/s41419-020-03088-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 08/13/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is a highly aggressive tumor with poor prognosis. A small subpopulation of glioma stem cells (GSCs) has been implicated in radiation resistance and tumor recurrence. In this study we analyzed the expression of miRNAs associated with the functions of GSCs using miRNA microarray analysis of these cells compared with human neural stem cells. These analyses identified gene clusters associated with glioma cell invasiveness, axonal guidance, and TGF-β signaling. miR-504 was significantly downregulated in GSCs compared with NSCs, its expression was lower in GBM compared with normal brain specimens and further decreased in the mesenchymal glioma subtype. Overexpression of miR-504 in GSCs inhibited their self-renewal, migration and the expression of mesenchymal markers. The inhibitory effect of miR-504 was mediated by targeting Grb10 expression which acts as an oncogene in GSCs and GBM. Overexpression of exogenous miR-504 resulted also in its delivery to cocultured microglia by GSC-secreted extracellular vesicles (EVs) and in the abrogation of the GSC-induced polarization of microglia to M2 subtype. Finally, miR-504 overexpression prolonged the survival of mice harboring GSC-derived xenografts and decreased tumor growth. In summary, we identified miRNAs and potential target networks that play a role in the stemness and mesenchymal transition of GSCs and the miR-504/Grb10 pathway as an important regulator of this process. Overexpression of miR-504 exerted antitumor effects in GSCs as well as bystander effects on the polarization of microglia via delivery by EVs.
Collapse
Affiliation(s)
- Ariel Bier
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Xin Hong
- Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Simona Cazacu
- Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Hodaya Goldstein
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Daniel Rand
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Cunli Xiang
- Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Wei Jiang
- Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Hiba Waldman Ben-Asher
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Moshe Attia
- Department of Neurosurgery, Sheba Medical Center, Henry Ford Hospital, Detroit, MI, USA
| | - Aharon Brodie
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ruicong She
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Laila M Poisson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Chaya Brodie
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
- Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
182
|
Zhang D, Xu X, Ye Q. Metabolism and immunity in breast cancer. Front Med 2020; 15:178-207. [PMID: 33074528 DOI: 10.1007/s11684-020-0793-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/17/2020] [Indexed: 12/12/2022]
Abstract
Breast cancer is one of the most common malignancies that seriously threaten women's health. In the process of the malignant transformation of breast cancer, metabolic reprogramming and immune evasion represent the two main fascinating characteristics of cancer and facilitate cancer cell proliferation. Breast cancer cells generate energy through increased glucose metabolism. Lipid metabolism contributes to biological signal pathways and forms cell membranes except energy generation. Amino acids act as basic protein units and metabolic regulators in supporting cell growth. For tumor-associated immunity, poor immunogenicity and heightened immunosuppression cause breast cancer cells to evade the host's immune system. For the past few years, the complex mechanisms of metabolic reprogramming and immune evasion are deeply investigated, and the genes involved in these processes are used as clinical therapeutic targets for breast cancer. Here, we review the recent findings related to abnormal metabolism and immune characteristics, regulatory mechanisms, their links, and relevant therapeutic strategies.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China.
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China.
| |
Collapse
|
183
|
Haddad AF, Chen JS, Oh T, Pereira MP, Joshi RS, Aghi MK. Higher cytolytic score correlates with an immunosuppressive tumor microenvironment and reduced survival in glioblastoma. Sci Rep 2020; 10:17580. [PMID: 33067480 PMCID: PMC7567862 DOI: 10.1038/s41598-020-73793-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/27/2020] [Indexed: 01/07/2023] Open
Abstract
Cytolytic score (CYT), calculated from mRNA expression levels of granzyme and perforin, positively correlates with CD8+ T cell infiltration/activity in a variety of cancers. Unlike other cancers, higher CYT has been associated with worse prognosis in glioblastoma (GBM). To address this discrepancy, we sought to investigate the relationship between CYT and immune checkpoint gene score (ICGscore), as well as their correlation with patient survival and tumor immune cell infiltration. Clinical and RNA-sequencing data for patients with newly diagnosed GBM were obtained from The Cancer Genome Atlas. Maximally-selected rank statistics was used to dichotomize subgroups. CIBERSORT was used to estimate abudence of immune cell-types. Spearman correlation was used to characterize the relationship between CYT and ICGscore. Kaplan-Meier curves were generated for survival analysis. Overall, 28/151 patients had high CYT. High CYT was associated with a mesenchymal subtype (p < 0.001) and worse survival (7.45 vs. 12.2 months, p < 0.001). There were no differences in patient demographics, IDH/MGMT mutation status, or treatment. On subgroup analysis, patients with high CYT/ICGscore had significantly increased CD8+ infiltration (p < 0.001), as expected, and worse survival (HR 0.445, p < 0.01). Furthermore, CYT strongly correlated with ICGscore (RS = 0.675, p < 0.001). The high CYT/ICGscore subgroup was associated with greater infiltration of M2 macrophages (p = 0.011) and neutrophils (p = 0.055). Our study highlights a multidimensional immunosuppressive GBM microenvironment in patients with higher CYT and potentially identifies patients with high CYT/ICGscore as a subgroup that may particularly benefit from multi-faceted immunotherapies, given their already elevated tumor CD8+ T cell levels.
Collapse
Affiliation(s)
- Alexander F Haddad
- School of Medicine, University of California San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143-0410, USA.
| | - Jia-Shu Chen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, USA
| | - Taemin Oh
- Department of Neurological Surgery, University of California San Francisco, San Francisco, USA
| | - Matheus P Pereira
- School of Medicine, University of California San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143-0410, USA
| | - Rushikesh S Joshi
- School of Medicine, University of California San Diego, San Diego, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, USA
| |
Collapse
|
184
|
López de Andrés J, Griñán-Lisón C, Jiménez G, Marchal JA. Cancer stem cell secretome in the tumor microenvironment: a key point for an effective personalized cancer treatment. J Hematol Oncol 2020; 13:136. [PMID: 33059744 PMCID: PMC7559894 DOI: 10.1186/s13045-020-00966-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) represent a tumor subpopulation responsible for tumor metastasis and resistance to chemo- and radiotherapy, ultimately leading to tumor relapse. As a consequence, the detection and eradication of this cell subpopulation represent a current challenge in oncology medicine. CSC phenotype is dependent on the tumor microenvironment (TME), which involves stem and differentiated tumor cells, as well as different cell types, such as mesenchymal stem cells, endothelial cells, fibroblasts and cells of the immune system, in addition to the extracellular matrix (ECM), different in composition to the ECM in healthy tissues. CSCs regulate multiple cancer hallmarks through the interaction with cells and ECM in their environment by secreting extracellular vesicles including exosomes, and soluble factors such as interleukins, cytokines, growth factors and other metabolites to the TME. Through these factors, CSCs generate and activate their own tumor niche by recruiting stromal cells and modulate angiogenesis, metastasis, resistance to antitumor treatments and their own maintenance by the secretion of different factors such as IL-6, VEGF and TGF-ß. Due to the strong influence of the CSC secretome on disease development, the new antitumor therapies focus on targeting these communication networks to eradicate the tumor and prevent metastasis, tumor relapse and drug resistance. This review summarizes for the first time the main components of the CSC secretome and how they mediate different tumor processes. Lastly, the relevance of the CSC secretome in the development of more precise and personalized antitumor therapies is discussed.
Collapse
Affiliation(s)
- Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain.,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain.,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain. .,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain. .,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain. .,Department of Health Sciences, University of Jaén, 23071, Jaén, Spain.
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain. .,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain. .,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain. .,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain.
| |
Collapse
|
185
|
Zhai Y, Li G, Li R, Chang Y, Feng Y, Wang D, Wu F, Zhang W. Single-Cell RNA-Sequencing Shift in the Interaction Pattern Between Glioma Stem Cells and Immune Cells During Tumorigenesis. Front Immunol 2020; 11:581209. [PMID: 33133100 PMCID: PMC7580180 DOI: 10.3389/fimmu.2020.581209] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is one of the most common neoplasms in the central nervous system characterized by limited immune response and unlimited expansion capability. Cancer stem cells (GSCs), a small fraction of the tumor cells, possess a pivotal regulation capability in the tumor microenvironment with a superior proliferation ability. We aimed to reveal the interaction between glioma stem cells (GSCs) and immune cells during tumorigenesis. Single-cell sequencing data from seven surgical specimens of glioblastoma patients and patient-derived GSCs cocultured with peripheral leukocytes were used for the analysis. Cell grouping and trajectory analysis were performed using Seurat and Monocle 3 packages in R software. The gene set of Cancer Genome Anatomy Project was used to define different cell types. Cells with the ability of proliferation and differentiation in glioblastoma tissue were defined as GSCs, which had a similar expression pattern to that in the GSCs in vitro. Astrocytes in glioblastoma were mainly derived from differentiated GSCs, while oligodendrocytes were most likely to be derived from different precursor cells. No remarkable evolutionary trajectory was observed among the subgroups of T cells in glioblastoma. The immune checkpoint interaction between GSCs and immune cells was changed from stimulatory to inhibitory during tumorigenesis. The patient-derived GSCs system is an ideal model for GSC research. The above research revealed that the interaction pattern between GSC glioma stem cells and immune cells during tumorigenesis provides a theoretical basis for GSC glioma stem cell-targeted immunotherapy.
Collapse
Affiliation(s)
- You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Renpeng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuemei Feng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network and Asian Glioma Genome Atlas Network, Beijing, China
| |
Collapse
|
186
|
De Boeck A, Ahn BY, D'Mello C, Lun X, Menon SV, Alshehri MM, Szulzewsky F, Shen Y, Khan L, Dang NH, Reichardt E, Goring KA, King J, Grisdale CJ, Grinshtein N, Hambardzumyan D, Reilly KM, Blough MD, Cairncross JG, Yong VW, Marra MA, Jones SJM, Kaplan DR, McCoy KD, Holland EC, Bose P, Chan JA, Robbins SM, Senger DL. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat Commun 2020; 11:4997. [PMID: 33020472 PMCID: PMC7536425 DOI: 10.1038/s41467-020-18569-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Despite a deeper molecular understanding, human glioblastoma remains one of the most treatment refractory and fatal cancers. It is known that the presence of macrophages and microglia impact glioblastoma tumorigenesis and prevent durable response. Herein we identify the dual function cytokine IL-33 as an orchestrator of the glioblastoma microenvironment that contributes to tumorigenesis. We find that IL-33 expression in a large subset of human glioma specimens and murine models correlates with increased tumor-associated macrophages/monocytes/microglia. In addition, nuclear and secreted functions of IL-33 regulate chemokines that collectively recruit and activate circulating and resident innate immune cells creating a pro-tumorigenic environment. Conversely, loss of nuclear IL-33 cripples recruitment, dramatically suppresses glioma growth, and increases survival. Our data supports the paradigm that recruitment and activation of immune cells, when instructed appropriately, offer a therapeutic strategy that switches the focus from the cancer cell alone to one that includes the normal host environment.
Collapse
Affiliation(s)
- Astrid De Boeck
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bo Young Ahn
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Charlotte D'Mello
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Xueqing Lun
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shyam V Menon
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mana M Alshehri
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Frank Szulzewsky
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Lubaba Khan
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ngoc Ha Dang
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Elliott Reichardt
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kimberly-Ann Goring
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer King
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cameron J Grisdale
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Natalie Grinshtein
- Department of Molecular Genetics, University of Toronto and Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute and the Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Karlyne M Reilly
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Michael D Blough
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - J Gregory Cairncross
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - David R Kaplan
- Department of Molecular Genetics, University of Toronto and Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Eric C Holland
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Pinaki Bose
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer A Chan
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Stephen M Robbins
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Donna L Senger
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
187
|
Martins TA, Schmassmann P, Shekarian T, Boulay JL, Ritz MF, Zanganeh S, Vom Berg J, Hutter G. Microglia-Centered Combinatorial Strategies Against Glioblastoma. Front Immunol 2020; 11:571951. [PMID: 33117364 PMCID: PMC7552736 DOI: 10.3389/fimmu.2020.571951] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated microglia (MG) and macrophages (MΦ) are important components of the glioblastoma (GBM) immune tumor microenvironment (iTME). From the recent advances in understanding how MG and GBM cells evolve and interact during tumorigenesis, we emphasize the cooperation of MG with other immune cell types of the GBM-iTME, mainly MΦ and T cells. We provide a comprehensive overview of current immunotherapeutic clinical trials and approaches for the treatment of GBM, which in general, underestimate the counteracting contribution of immunosuppressive MG as a main factor for treatment failure. Furthermore, we summarize new developments and strategies in MG reprogramming/re-education in the GBM context, with a focus on ways to boost MG-mediated tumor cell phagocytosis and associated experimental models and methods. This ultimately converges in our proposal of novel combinatorial regimens that locally modulate MG as a central paradigm, and therefore may lead to additional, long-lasting, and effective tumoricidal responses.
Collapse
Affiliation(s)
- Tomás A Martins
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Tala Shekarian
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jean-Louis Boulay
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Marie-Françoise Ritz
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Steven Zanganeh
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Chemical and Biomolecular Engineering, New York University, New York, NY, United States
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
| | - Gregor Hutter
- Department of Biomedicine, University of Basel, Basel, Switzerland.,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
188
|
Lin X, Wang Z, Yang G, Wen G, Zhang H. YTHDF2 correlates with tumor immune infiltrates in lower-grade glioma. Aging (Albany NY) 2020; 12:18476-18500. [PMID: 32986017 PMCID: PMC7585119 DOI: 10.18632/aging.103812] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/20/2020] [Indexed: 01/24/2023]
Abstract
Immunotherapy is an effective treatment for many cancer types. However, YTHDF2 effects on the prognosis of different tumors and correlation with tumor immune infiltration are unclear. Here, we analyzed The Cancer Genome Atlas and Gene Expression Omnibus data obtained through various web-based platforms. The analyses showed that YTHDF2 expression and associated prognoses may depend on cancer type. High YTHDF2 expression was associated with poor overall survival in lower-grade glioma (LGG). In addition, YTHDF2 expression positively correlated with expression of several immune cell markers, including PD-1, TIM-3, and CTLA-4, as well as tumor-associated macrophage gene markers, and isocitrate dehydrogenase 1 in LGG. These findings suggest that YTHDF2 is a potential prognostic biomarker that correlates with LGG tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Xiangan Lin
- Department of Cancer Chemotherapy, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510000, China
| | - Zhichao Wang
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| | - Guangda Yang
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| | - Guohua Wen
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| | - Hailiang Zhang
- Department of Cancer Chemotherapy, Zengcheng District People’s Hospital of Guangzhou, Guangzhou 511300, China
| |
Collapse
|
189
|
Kinoh H, Quader S, Shibasaki H, Liu X, Maity A, Yamasoba T, Cabral H, Kataoka K. Translational Nanomedicine Boosts Anti-PD1 Therapy to Eradicate Orthotopic PTEN-Negative Glioblastoma. ACS NANO 2020; 14:10127-10140. [PMID: 32806051 DOI: 10.1021/acsnano.0c03386] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Glioblastoma (GBM) is resistant to immune checkpoint inhibition due to its low mutation rate, phosphatase and tensin homologue (PTEN)-deficient immunosuppressive microenvironment, and high fraction of cancer stem-like cells (CSCs). Nanomedicines fostering immunoactivating intratumoral signals could reverse GBM resistance to immune checkpoint inhibitors (ICIs) for promoting curative responses. Here, we applied pH-sensitive epirubicin-loaded micellar nanomedicines, which are under clinical evaluation, to synergize the efficacy of anti-PD1antibodies (aPD1) against PTEN-positive and PTEN-negative orthotopic GBM, the latter with a large subpopulation of CSCs. The combination of epirubicin-loaded micelles (Epi/m) with aPD1 overcame GBM resistance to ICIs by transforming cold GBM into hot tumors with high infiltration of antitumor immune cells through the induction of immunogenic cell death (ICD), elimination of immunosuppressive myeloid-derived suppressor cells (MSDCs), and reduction of PD-L1 expression on tumor cells. Thus, Epi/m plus aPD1 eradicated both PTEN-positive and PTEN-negative orthotopic GBM and provided long-term immune memory effects. Our results indicate the high translatable potential of Epi/m plus aPD1 for the treatment of GBM.
Collapse
Affiliation(s)
- Hiroaki Kinoh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Hitoshi Shibasaki
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Otolaryngology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Xueying Liu
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Amit Maity
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Tatsuya Yamasoba
- Department of Otolaryngology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
190
|
Dumas AA, Pomella N, Rosser G, Guglielmi L, Vinel C, Millner TO, Rees J, Aley N, Sheer D, Wei J, Marisetty A, Heimberger AB, Bowman RL, Brandner S, Joyce JA, Marino S. Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J 2020; 39:e103790. [PMID: 32567735 PMCID: PMC7396846 DOI: 10.15252/embj.2019103790] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 05/03/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022] Open
Abstract
Tumour-associated microglia/macrophages (TAM) are the most numerous non-neoplastic populations in the tumour microenvironment in glioblastoma multiforme (GBM), the most common malignant brain tumour in adulthood. The mTOR pathway, an important regulator of cell survival/proliferation, is upregulated in GBM, but little is known about the potential role of this pathway in TAM. Here, we show that GBM-initiating cells induce mTOR signalling in the microglia but not bone marrow-derived macrophages in both in vitro and in vivo GBM mouse models. mTOR-dependent regulation of STAT3 and NF-κB activity promotes an immunosuppressive microglial phenotype. This hinders effector T-cell infiltration, proliferation and immune reactivity, thereby contributing to tumour immune evasion and promoting tumour growth in mouse models. The translational value of our results is demonstrated in whole transcriptome datasets of human GBM and in a novel in vitro model, whereby expanded-potential stem cells (EPSC)-derived microglia-like cells are conditioned by syngeneic patient-derived GBM-initiating cells. These results raise the possibility that microglia could be the primary target of mTOR inhibition, rather than the intrinsic tumour cells in GBM.
Collapse
Affiliation(s)
- Anaelle A Dumas
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Nicola Pomella
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Gabriel Rosser
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Loredana Guglielmi
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Claire Vinel
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Thomas O Millner
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Jeremy Rees
- National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Natasha Aley
- Division of NeuropathologyDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - Denise Sheer
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Jun Wei
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Anantha Marisetty
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Amy B Heimberger
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Robert L Bowman
- Human Oncology and Pathogenesis ProgramMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Sebastian Brandner
- National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Johanna A Joyce
- Department of OncologyLudwig Institute for Cancer ResearchUniversity of LausanneLausanneSwitzerland
| | - Silvia Marino
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| |
Collapse
|
191
|
Birch JL, Coull BJ, Spender LC, Watt C, Willison A, Syed N, Chalmers AJ, Hossain-Ibrahim MK, Inman GJ. Multifaceted transforming growth factor-beta (TGFβ) signalling in glioblastoma. Cell Signal 2020; 72:109638. [PMID: 32320860 DOI: 10.1016/j.cellsig.2020.109638] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is an aggressive and devastating primary brain cancer which responds very poorly to treatment. The average survival time of patients is only 14-15 months from diagnosis so there is a clear and unmet need for the development of novel targeted therapies to improve patient outcomes. The multifunctional cytokine TGFβ plays fundamental roles in development, adult tissue homeostasis, tissue wound repair and immune responses. Dysfunction of TGFβ signalling has been implicated in both the development and progression of many tumour types including GBM, thereby potentially providing an actionable target for its treatment. This review will examine TGFβ signalling mechanisms and their role in the development and progression of GBM. The targeting of TGFβ signalling using a variety of approaches including the TGFβ binding protein Decorin will be highlighted as attractive therapeutic strategies.
Collapse
Affiliation(s)
| | - Barry J Coull
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Lindsay C Spender
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Courtney Watt
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Alice Willison
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Nelofer Syed
- The John Fulcher Molecular Neuro-Oncology Laboratory, Division of Brain Sciences, Imperial College London, London, UK
| | | | - M Kismet Hossain-Ibrahim
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK; Department of Neurosurgery, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK
| | - Gareth J Inman
- CRUK Beatson Institute, Glasgow, UK; Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
192
|
DeCordova S, Shastri A, Tsolaki AG, Yasmin H, Klein L, Singh SK, Kishore U. Molecular Heterogeneity and Immunosuppressive Microenvironment in Glioblastoma. Front Immunol 2020; 11:1402. [PMID: 32765498 PMCID: PMC7379131 DOI: 10.3389/fimmu.2020.01402] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, with a poor prognosis, despite surgical resection combined with radio- and chemotherapy. The major clinical obstacles contributing to poor GBM prognosis are late diagnosis, diffuse infiltration, pseudo-palisading necrosis, microvascular proliferation, and resistance to conventional therapy. These challenges are further compounded by extensive inter- and intra-tumor heterogeneity and the dynamic plasticity of GBM cells. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumor microenvironment, which mostly induces tumor aggressiveness and drug resistance. An immunosuppressive tumor microenvironment of GBM provides multiple pathways for tumor immune evasion. Infiltrating immune cells, mostly tumor-associated macrophages, comprise much of the non-neoplastic population in GBM. Further understanding of the immune microenvironment of GBM is essential to make advances in the development of immunotherapeutics. Recently, whole-genome sequencing, epigenomics and transcriptional profiling have significantly helped improve the prognostic and therapeutic outcomes of GBM patients. Here, we discuss recent genomic advances, the role of innate and adaptive immune mechanisms, and the presence of an established immunosuppressive GBM microenvironment that suppresses and/or prevents the anti-tumor host response.
Collapse
Affiliation(s)
- Syreeta DeCordova
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Abhishek Shastri
- Central and North West London NHS Foundation Trust, London, United Kingdom
| | - Anthony G Tsolaki
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, India
| | - Lukas Klein
- Department of Gastroenterology and Gastroenterology Oncology, University Medical Centre, Göttingen, Germany
| | - Shiv K Singh
- Department of Gastroenterology and Gastroenterology Oncology, University Medical Centre, Göttingen, Germany
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| |
Collapse
|
193
|
Macrophages in Oral Carcinomas: Relationship with Cancer Stem Cell Markers and PD-L1 Expression. Cancers (Basel) 2020; 12:cancers12071764. [PMID: 32630659 PMCID: PMC7408350 DOI: 10.3390/cancers12071764] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor-associated macrophages (TAMs) can be polarized into antitumoral M1 and protumoral and immunosuppressive M2 macrophages. This study investigated the clinical relevance of TAM infiltration in oral squamous cell carcinoma (OSCC), evaluating CD68 (M1 and M2 macrophage marker) and CD163 expression (M2 macrophage marker) in the tumor nests and surrounding stroma. Immunohistochemical analysis of both stromal/tumoral CD68+ and CD163+ TAMs was performed in paraffin-embedded tissue specimens from 125 OSCC patients, and correlated with clinical data. Potential relationships with the expression of cancer stem cell (CSC) markers and PD-L1 in the tumors were also assessed. Stromal CD163+ infiltration was significantly associated with the tumor location in the tongue, and stromal and tumoral CD68+ and CD163+-infiltrating TAMs were more abundant in nonsmokers and non-alcohol-drinkers. Strikingly, this study uncovers an inverse relationship between CD68+ and CD163+ TAMs and CSC marker expression (NANOG and SOX2) in OSCC. High infiltration of CD163+ TAMs in both tumor and stroma was strongly and significantly correlated with the absence of NANOG expression. Moreover, infiltration of both CD68+ and CD163+ TAMs was also significantly associated with high tumor expression of PD-L1. Our results suggest that there is a link between TAM infiltration and immune escape in OSCC.
Collapse
|
194
|
Ott M, Kassab C, Marisetty A, Hashimoto Y, Wei J, Zamler D, Leu JS, Tomaszowski KH, Sabbagh A, Fang D, Gupta P, Priebe W, Zielinski RJ, Burks JK, Long JP, Kong LY, Fuller GN, DeGroot J, Sulman EP, Heimberger AB. Radiation with STAT3 Blockade Triggers Dendritic Cell-T cell Interactions in the Glioma Microenvironment and Therapeutic Efficacy. Clin Cancer Res 2020; 26:4983-4994. [PMID: 32605912 DOI: 10.1158/1078-0432.ccr-19-4092] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/14/2020] [Accepted: 06/24/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with central nervous system (CNS) tumors are typically treated with radiotherapy, but this is not curative and results in the upregulation of phosphorylated STAT3 (p-STAT3), which drives invasion, angiogenesis, and immune suppression. Therefore, we investigated the combined effect of an inhibitor of STAT3 and whole-brain radiotherapy (WBRT) in a murine model of glioma. EXPERIMENTAL DESIGN C57BL/6 mice underwent intracerebral implantation of GL261 glioma cells, WBRT, and treatment with WP1066, a blood-brain barrier-penetrant inhibitor of the STAT3 pathway, or the two in combination. The role of the immune system was evaluated using tumor rechallenge strategies, immune-incompetent backgrounds, immunofluorescence, immune phenotyping of tumor-infiltrating immune cells (via flow cytometry), and NanoString gene expression analysis of 770 immune-related genes from immune cells, including those directly isolated from the tumor microenvironment. RESULTS The combination of WP1066 and WBRT resulted in long-term survivors and enhanced median survival time relative to monotherapy in the GL261 glioma model (combination vs. control P < 0.0001). Immunologic memory appeared to be induced, because mice were protected during subsequent tumor rechallenge. The therapeutic effect of the combination was completely lost in immune-incompetent animals. NanoString analysis and immunofluorescence revealed immunologic reprograming in the CNS tumor microenvironment specifically affecting dendritic cell antigen presentation and T-cell effector functions. CONCLUSIONS This study indicates that the combination of STAT3 inhibition and WBRT enhances the therapeutic effect against gliomas in the CNS by inducing dendritic cell and T-cell interactions in the CNS tumor.
Collapse
Affiliation(s)
- Martina Ott
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia Kassab
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anantha Marisetty
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuuri Hashimoto
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jun Wei
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Zamler
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jia-Shiun Leu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Karl-Heinz Tomaszowski
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aria Sabbagh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dexing Fang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pravesh Gupta
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rafal J Zielinski
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Department of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John DeGroot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erik P Sulman
- Department of Radiation Oncology, NYU Langone Health Perlmutter Cancer Center, New York, New York
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
195
|
Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma. Nat Commun 2020; 11:3015. [PMID: 32541784 PMCID: PMC7295765 DOI: 10.1038/s41467-020-16827-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
The interplay between glioma stem cells (GSCs) and the tumor microenvironment plays crucial roles in promoting malignant growth of glioblastoma (GBM), the most lethal brain tumor. However, the molecular mechanisms underlying this crosstalk are incompletely understood. Here, we show that GSCs secrete the Wnt‐induced signaling protein 1 (WISP1) to facilitate a pro-tumor microenvironment by promoting the survival of both GSCs and tumor-associated macrophages (TAMs). WISP1 is preferentially expressed and secreted by GSCs. Silencing WISP1 markedly disrupts GSC maintenance, reduces tumor-supportive TAMs (M2), and potently inhibits GBM growth. WISP1 signals through Integrin α6β1-Akt to maintain GSCs by an autocrine mechanism and M2 TAMs through a paracrine manner. Importantly, inhibition of Wnt/β-catenin-WISP1 signaling by carnosic acid (CA) suppresses GBM tumor growth. Collectively, these data demonstrate that WISP1 plays critical roles in maintaining GSCs and tumor-supportive TAMs in GBM, indicating that targeting Wnt/β-catenin-WISP1 signaling may effectively improve GBM treatment and the patient survival. The tumour microenvironment plays an important role in promoting glioblastoma. Here, the authors show that glioma stem cells secrete WISP1, which promotes both the survival of the stem cells and tumour-associated macrophages.
Collapse
|
196
|
Huang L, Wang Z, Chang Y, Wang K, Kang X, Huang R, Zhang Y, Chen J, Zeng F, Wu F, Zhao Z, Li G, Huang H, Jiang T, Hu H. EFEMP2 indicates assembly of M0 macrophage and more malignant phenotypes of glioma. Aging (Albany NY) 2020; 12:8397-8412. [PMID: 32396873 PMCID: PMC7244085 DOI: 10.18632/aging.103147] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 02/19/2020] [Indexed: 05/11/2023]
Abstract
Immune response mediated by macrophages is critical in tumor progression and implicates new targets in potential efficient immunotherapies. Tumor associated macrophages (TAM) are divided into either polarized M1 or M2 phenotype depending on different regulators of polarization and pro- or anti-oncogenic roles they play. Glioma-infiltrated TAMs have been newly reported contrary to the current polarization dogma. Instead, macrophages in glioma exhibit a continuum phenotype between the M1- and M2-like TAM that resembling M0 macrophage. Here we proposed an OS (overall survival)-correlated gene EFEMP2 (EGF containing fibulin-like extracellular matrix protein 2) via screening with transcriptional expression levels and methylation data in two glioma databases. EFEMP2 was found highly expressed in glioma of higher WHO grade and Mesenchymal subtype glioma, and its transcriptional level could predict OS efficiently in validation datasets. EFEMP2 exhibited a remarkable preference of intercellular expression. In vitro assay showed that EFEMP2's level in medium was closely related to glioma cells' growth. Moreover, EFEMP2 expression level was remarkably correlated with immunological responses. M0-like macrophage as a feature of malignancy of glioblastoma revealed distinct assembly in glioma with high level of EFEMP2. These results revealed EFEMP2's role as a potential characteristic marker of malignant glioma, which are enriched of M0 macrophage.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Zheng Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Yuanhao Chang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Kuanyu Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Xun Kang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Jing Chen
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Hua Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| |
Collapse
|
197
|
Muir M, Gopakumar S, Traylor J, Lee S, Rao G. Glioblastoma multiforme: novel therapeutic targets. Expert Opin Ther Targets 2020; 24:605-614. [PMID: 32394767 DOI: 10.1080/14728222.2020.1762568] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The increasingly detailed genetic characterization of glioblastoma (GBM) has failed to translate into meaningful breakthroughs in treatment. This is likely to be attributed to molecular heterogeneity of GBM. However, the understanding of the tumor microenvironment in GBM has become more refined and has revealed a wealth of therapeutic targets that may enable the disruption of angiogenesis or immunosuppression. AREAS COVERED This review discusses the selective targeting of tumor-intrinsic pathways, therapies that target the GBM tumor microenvironment and relevant preclinical studies and their limitations. Relevant literature was derived from a PubMed search encompassing studies from 1989 to 2020. EXPERT OPINION Despite appropriate target engagement, attempts to directly inhibit oncogenic pathways in GBM have yielded little success. This is likely attributed to the molecular heterogeneity of GBM and the presence of redundant signaling that allow for accumulation of adaptive mutations and development of drug resistance. Subsequently, there has been a shift toward therapies modulating the pro-angiogenic, immunosuppressive tumor microenvironment in GBM. The non-transformed cells in the microenvironment which includes endothelial cells, myeloid cells, and T cells, are presumably genetically stable, less susceptible to heterogeneity, and easier to target. This approach offers the highest potential for a therapeutic breakthrough in GBM.
Collapse
Affiliation(s)
- Matthew Muir
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX, USA
| | | | - Jeffrey Traylor
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX, USA
| | - Sungho Lee
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX, USA
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX, USA.,Department of Neurosurgery, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
198
|
Aaliyari-Serej Z, Ebrahimi A, Barazvan B, Ebrahimi-Kalan A, Hajiasgharzadeh K, Kazemi T, Baradaran B. Recent Advances in Targeting of Breast Cancer Stem Cells Based on Biological Concepts and Drug Delivery System Modification. Adv Pharm Bull 2020; 10:338-349. [PMID: 32665892 PMCID: PMC7335982 DOI: 10.34172/apb.2020.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer with various biological diversity known as the common reason of death in the world and despite progress in novel therapeutic approaches, it faced with failure and recurrence in general. Recent clinical and preclinical statistics support cancer stem cells (CSCs) hypothesis and its similarities with normal stem cells. Evaluation of related paper conclude in significance finding in the further characterization of CSCs biology such as surface biomarkers, microenvironment regulatory molecules, cell signaling pathways, cell to cell transition and drug efflux pumps to overcome multidrug resistance and effective therapy. Emerging novel data indicate biological concepts in the base of unsuccessful treatment. A powerful understanding of the cell signaling pathways in cancer and CSCs topics can be led us to define and control treatment problems in cancer. More recently nano medicine based on drug delivery system modification and new implications on combinatorial therapy have been used to treat breast cancer effectively. The aim of this review is focus on CSCs as a potential target of cancer therapy, to overcome the limitation and problems of current therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Zeynab Aaliyari-Serej
- Department of Applied Cell Sciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayyub Ebrahimi
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Halic Uuniversity, Istanbul, Turkey
| | - Balal Barazvan
- Department of Basic Sciences, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
199
|
Pastò A, Consonni FM, Sica A. Influence of Innate Immunity on Cancer Cell Stemness. Int J Mol Sci 2020; 21:ijms21093352. [PMID: 32397392 PMCID: PMC7247585 DOI: 10.3390/ijms21093352] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Even if cancer stem cells (CSCs) represent only a small proportion of the tumor mass, they significantly account for tumor maintenance, resistance to therapies, relapse and metastatic spread, due to their increased capacity of self-renewal, multipotency, tumorigenicity and quiescence. Emerging evidence suggests that the immune contexture within the tumor microenvironment (TME) determines both the response to therapy and the clinical outcome. In this context, CSCs acquire immune evasion skills by editing immune cell functions and sculpting the immunosuppressive landscape of TME. Reciprocally, infiltrating immune cells influence CSCs self-renewal, tumorigenicity and metastasis. In this review, we summarize the immunomodulatory properties of CSCs, as well as the impact of innate immune cells on cancer cells stemness in the different phases of cancer immunoediting process and neoplastic progression.
Collapse
Affiliation(s)
- Anna Pastò
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
| | - Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
| | - Antonio Sica
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
- Correspondence: ; Tel.: +39-0321-375-881; Fax: +39-0321-375-621
| |
Collapse
|
200
|
Rajapakse VN, Herrada S, Lavi O. Phenotype stability under dynamic brain-tumor environment stimuli maps glioblastoma progression in patients. SCIENCE ADVANCES 2020; 6:eaaz4125. [PMID: 32832595 PMCID: PMC7439317 DOI: 10.1126/sciadv.aaz4125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/20/2020] [Indexed: 06/11/2023]
Abstract
Although tumor invasiveness is known to drive glioblastoma (GBM) recurrence, current approaches to treatment assume a fairly simple GBM phenotype transition map. We provide new analyses to estimate the likelihood of reaching or remaining in a phenotype under dynamic, physiologically likely perturbations of stimuli ("phenotype stability"). We show that higher stability values of the motile phenotype (Go) are associated with reduced patient survival. Moreover, induced motile states are capable of driving GBM recurrence. We found that the Dormancy and Go phenotypes are equally represented in advanced GBM samples, with natural transitioning between the two. Furthermore, Go and Grow phenotype transitions are mostly driven by tumor-brain stimuli. These are difficult to regulate directly, but could be modulated by reprogramming tumor-associated cell types. Our framework provides a foundation for designing targeted perturbations of the tumor-brain environment, by assessing their impact on GBM phenotypic plasticity, and is corroborated by analyses of patient data.
Collapse
Affiliation(s)
- Vinodh N. Rajapakse
- Integrative Cancer Dynamics Unit, Laboratory of Cell Biology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Sylvia Herrada
- Integrative Cancer Dynamics Unit, Laboratory of Cell Biology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Orit Lavi
- Integrative Cancer Dynamics Unit, Laboratory of Cell Biology, CCR, NCI, NIH, Bethesda, MD, USA
| |
Collapse
|