401
|
Reardon DA, Freeman G, Wu C, Chiocca EA, Wucherpfennig KW, Wen PY, Fritsch EF, Curry WT, Sampson JH, Dranoff G. Immunotherapy advances for glioblastoma. Neuro Oncol 2014; 16:1441-58. [PMID: 25190673 DOI: 10.1093/neuonc/nou212] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Survival for patients with glioblastoma, the most common high-grade primary CNS tumor, remains poor despite multiple therapeutic interventions including intensifying cytotoxic therapy, targeting dysregulated cell signaling pathways, and blocking angiogenesis. Exciting, durable clinical benefits have recently been demonstrated for a number of other challenging cancers using a variety of immunotherapeutic approaches. Much modern research confirms that the CNS is immunoactive rather than immunoprivileged. Preliminary results of clinical studies demonstrate that varied vaccine strategies have achieved encouraging evidence of clinical benefit for glioblastoma patients, although multiple variables will likely require systematic investigation before optimal outcomes are realized. Initial preclinical studies have also revealed promising results with other immunotherapies including cell-based approaches and immune checkpoint blockade. Clinical studies to evaluate a wide array of immune therapies for malignant glioma patients are being rapidly developed. Important considerations going forward include optimizing response assessment and identifiying correlative biomarkers for predict therapeutic benefit. Finally, the potential of complementary combinatorial immunotherapeutic regimens is highly exciting and warrants expedited investigation.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Gordon Freeman
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Catherine Wu
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - E Antonio Chiocca
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Kai W Wucherpfennig
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Edward F Fritsch
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - William T Curry
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - John H Sampson
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Glenn Dranoff
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| |
Collapse
|
402
|
Xu S, Wei J, Wang F, Kong LY, Ling XY, Nduom E, Gabrusiewicz K, Doucette T, Yang Y, Yaghi NK, Fajt V, Levine JM, Qiao W, Li XG, Lang FF, Rao G, Fuller GN, Calin GA, Heimberger AB. Effect of miR-142-3p on the M2 macrophage and therapeutic efficacy against murine glioblastoma. J Natl Cancer Inst 2014; 106:dju162. [PMID: 24974128 DOI: 10.1093/jnci/dju162] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The immune therapeutic potential of microRNAs (miRNAs) in the context of tumor-mediated immune suppression has not been previously described for monocyte-derived glioma-associated macrophages, which are the largest infiltrating immune cell population in glioblastomas and facilitate gliomagenesis. METHODS An miRNA microarray was used to compare expression profiles between human glioblastoma-infiltrating macrophages and matched peripheral monocytes. The effects of miR-142-3p on phenotype and function of proinflammatory M1 and immunosuppressive M2 macrophages were determined. The therapeutic effect of miR-142-3p was ascertained in immune-competent C57BL/6J mice harboring intracerebral GL261 gliomas and in genetically engineered Ntv-a mice bearing high-grade gliomas. Student t test was used to evaluate the differences between ex vivo datasets. Survival was analyzed with the log-rank test and tumor sizes with linear mixed models and F test. All statistical tests were two-sided. RESULTS miR-142-3p was the most downregulated miRNA (approximately 4.95-fold) in glioblastoma-infiltrating macrophages. M2 macrophages had lower miR-142-3p expression relative to M1 macrophages (P = .03). Overexpression of miR-142-3p in M2 macrophages induced selective modulation of transforming growth factor beta receptor 1, which led to subsequent preferential apoptosis in the M2 subset (P = .01). In vivo miR-142-3p administration resulted in glioma growth inhibition (P = .03, n = 5) and extended median survival (miR-142-3p-treated C57BL/6J mice vs scramble control: 31 days vs 23.5 days, P = .03, n = 10; miR-142-3p treated Ntv-a mice vs scramble control: 32 days vs 24 days, P = .03, n = 9), with an associated decrease in infiltrating macrophages (R (2) = .303). CONCLUSIONS These data indicate a unique role of miR-142-3p in glioma immunity by modulating M2 macrophages through the transforming growth factor beta signaling pathway.
Collapse
Affiliation(s)
- Shuo Xu
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Jun Wei
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Fei Wang
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Ling-Yuan Kong
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Xiao-Yang Ling
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Edjah Nduom
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Konrad Gabrusiewicz
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Tiffany Doucette
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Yuhui Yang
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Nasser K Yaghi
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Virginia Fajt
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Jonathan M Levine
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Wei Qiao
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Xin-Gang Li
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Frederick F Lang
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Ganesh Rao
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Gregory N Fuller
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - George A Calin
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF)
| | - Amy B Heimberger
- Affiliations of authors: Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (SX, X-GL), Department of Neurosurgery (SX, JW, FW, L-YK, X-YL, EN, KG, TD, YY, FFL, GR, ABH), Department of Biostatistics (WQ), Department of Pathology (GNF), and Department of Experimental Therapeutics (GAC), University of Texas M. D. Anderson Cancer Center, Houston, TX; Baylor College of Medicine, Houston, TX (NKY); Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX (VF).
| |
Collapse
|
403
|
Rébé C, Végran F, Berger H, Ghiringhelli F. STAT3 activation: A key factor in tumor immunoescape. JAKSTAT 2014; 2:e23010. [PMID: 24058791 PMCID: PMC3670267 DOI: 10.4161/jkst.23010] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/21/2012] [Accepted: 11/27/2012] [Indexed: 12/28/2022] Open
Abstract
Cancer growth is controlled by cancer cells (cell intrinsic phenomenon), but also by the immune cells in the tumor microenvironment (cell extrinsic phenomenon). Thus cancer progression is mediated by the activation of transcription programs responsible for cancer cell proliferation, but also induced proliferation/activation of immunosuppressive cells such as Th17, Treg or myeloid derived suppressor cells (MDSCs). One of the key transcription factors involved in these pathways is the signal transducer and activator of transcription 3 (STAT3). In this review we will focus on STAT3 activation in immune cells, and how it impacts on tumor progression.
Collapse
Affiliation(s)
- Cédric Rébé
- INSERM, U866; Dijon, France ; Centre Georges François Leclerc; Dijon, France
| | | | | | | |
Collapse
|
404
|
Fu R, Shen Q, Xu P, Luo JJ, Tang Y. Phagocytosis of microglia in the central nervous system diseases. Mol Neurobiol 2014; 49:1422-34. [PMID: 24395130 PMCID: PMC4012154 DOI: 10.1007/s12035-013-8620-6] [Citation(s) in RCA: 440] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/15/2013] [Indexed: 12/20/2022]
Abstract
Microglia, the resident macrophages of the central nervous system, rapidly activate in nearly all kinds of neurological diseases. These activated microglia become highly motile, secreting inflammatory cytokines, migrating to the lesion area, and phagocytosing cell debris or damaged neurons. During the past decades, the secretory property and chemotaxis of microglia have been well-studied, while relatively less attention has been paid to microglial phagocytosis. So far there is no obvious concordance with whether it is beneficial or detrimental in tissue repair. This review focuses on phagocytic phenotype of microglia in neurological diseases such as Alzheimer's disease, multiple sclerosis, Parkinson's disease, traumatic brain injury, ischemic and other brain diseases. Microglial morphological characteristics, involved receptors and signaling pathways, distribution variation along with time and space changes, and environmental factors that affecting phagocytic function in each disease are reviewed. Moreover, a comparison of contributions between macrophages from peripheral circulation and the resident microglia to these pathogenic processes will also be discussed.
Collapse
Affiliation(s)
- Ruying Fu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
- Department of Neurology, Zengcheng People’s Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pengfei Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
| | - Jin Jun Luo
- Department of Neurology, School of Medicine, Temple University, Philadelphia, PA USA
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
405
|
Hou YC, Chao YJ, Tung HL, Wang HC, Shan YS. Coexpression of CD44-positive/CD133-positive cancer stem cells and CD204-positive tumor-associated macrophages is a predictor of survival in pancreatic ductal adenocarcinoma. Cancer 2014; 120:2766-77. [PMID: 24839953 PMCID: PMC4232049 DOI: 10.1002/cncr.28774] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/23/2014] [Accepted: 04/08/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND The interactions between cancer stem cells (CSCs) and tumor-associated macrophages (TAMs) can promote tumor progression, maintain the CSCs population, and reduce therapeutic effects. The objective of this study was to investigate the coexpression of CSCs and TAMs and its clinical significance in pancreatic ductal adenocarcinoma (PDAC). METHODS Ninety-six patients with PDAC were included in this study. Tissue microarrays were constructed for immunostaining of the CSCs markers CD44 and CD133 and the TAMs marker CD204. Correlations between the expression of CSCs and TAMs markers and clinicopathologic characteristics or disease progression were analyzed. RESULTS Expression levels of CD44/CD133 and CD204 were significantly higher in tumor tissues than in normal tissues (P < .0001). The variables associated with survival were high coexpression of CD44/CD133 (P = .000), high expression of CD204 (P = .011), and tumor grade (P = .014). There was a positive correlation between CD44/CD133 and CD204 expression (r = 0.294; P = .004). Survival analysis indicated that high coexpression of CD44/CD133 and CD204 was associated significantly with shorter overall survival (P = .000) and disease-free survival (P = .003). Multivariate analysis revealed that high CD44/CD133 expression was an independent prognostic factor for disease-free survival, whereas high CD204 expression was an independent predictor for both overall and disease-free survival. CONCLUSIONS Coexpression of CD44/CD133 and CD204 is a useful survival prediction marker for patients with PDAC. Cancer 2014;120:2766–2777. © The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. The clinical significance of pancreatic cancer stem cells and tumor-associated macrophages is explored in patients with pancreatic ductal adenocarcinoma. The results clearly demonstrate that coexpression of 2 cancer stem cell markers (CD44 and CD133) and a tumor-associated macrophage marker (CD204) is a useful prognostic factor for predicting the survival of patients with pancreatic ductal adenocarcinoma after surgery.
Collapse
Affiliation(s)
- Ya-Chin Hou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
406
|
Xiong A, Yang Z, Shen Y, Zhou J, Shen Q. Transcription Factor STAT3 as a Novel Molecular Target for Cancer Prevention. Cancers (Basel) 2014; 6:926-57. [PMID: 24743778 PMCID: PMC4074810 DOI: 10.3390/cancers6020926] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/11/2014] [Accepted: 03/18/2014] [Indexed: 12/11/2022] Open
Abstract
Signal Transducers and Activators of Transcription (STATs) are a family of transcription factors that regulate cell proliferation, differentiation, apoptosis, immune and inflammatory responses, and angiogenesis. Cumulative evidence has established that STAT3 has a critical role in the development of multiple cancer types. Because it is constitutively activated during disease progression and metastasis in a variety of cancers, STAT3 has promise as a drug target for cancer therapeutics. Recently, STAT3 was found to have an important role in maintaining cancer stem cells in vitro and in mouse tumor models, suggesting STAT3 is integrally involved in tumor initiation, progression and maintenance. STAT3 has been traditionally considered as nontargetable or undruggable, and the lag in developing effective STAT3 inhibitors contributes to the current lack of FDA-approved STAT3 inhibitors. Recent advances in cancer biology and drug discovery efforts have shed light on targeting STAT3 globally and/or specifically for cancer therapy. In this review, we summarize current literature and discuss the potential importance of STAT3 as a novel target for cancer prevention and of STAT3 inhibitors as effective chemopreventive agents.
Collapse
Affiliation(s)
- Ailian Xiong
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zhengduo Yang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yicheng Shen
- College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Qiang Shen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
407
|
Zhou W, Bao S. Reciprocal Supportive Interplay between Glioblastoma and Tumor-Associated Macrophages. Cancers (Basel) 2014; 6:723-40. [PMID: 24675569 PMCID: PMC4074800 DOI: 10.3390/cancers6020723] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal and aggressive type of primary brain malignancy. Failures of the traditional therapies in treating GBMs raise the urgent requirement to develop new approaches with more responsive targets. The phenomenon of the high infiltration of tumor-associated macrophages (TAMs) into GBMs has been observed for a long time. Regardless of the limited knowledge about TAMs, the high percentage of supportive TAM in GBM tumor mass makes it possible to be a good target for GBM treatment. In this review, we discussed the unique features of TAMs in GBMs, including their origin, the tumor-supportive properties, the secreted cytokines, and the relevant mechanisms. In addition, we tried to interpret the current understandings about the interplay between GBM cancer cells and TAMs. Finally, the translational studies of targeting TAMs were also described.
Collapse
Affiliation(s)
- Wenchao Zhou
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
408
|
Sarkar S, Yong VW. The battle for the brain: Brain tumor-initiating cells vs. microglia/macrophages. Oncoimmunology 2014; 3:e28047. [PMID: 25340004 PMCID: PMC4203533 DOI: 10.4161/onci.28047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 01/29/2014] [Indexed: 11/19/2022] Open
Abstract
Brain tumor-initiating cells (BTICs) become less tumorigenic when co-cultured with microglia/macrophages (M/Ms) isolated from subjects not affected by glioma, but not when exposed to the M/Ms of glioma patients. Microglial cells and macrophages from glioma patients, however, can be reactivated by non-toxic doses of amphotericin B to curb the growth of BTICs in vitro and in vivo.
Collapse
Affiliation(s)
- Susobhan Sarkar
- Hotchkiss Brain Institute and Department of Clinical Neurosciences; University of Calgary; Calgary, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences; University of Calgary; Calgary, Canada
| |
Collapse
|
409
|
Reardon DA, Wucherpfennig KW, Freeman G, Wu CJ, Chiocca EA, Wen PY, Curry WT, Mitchell DA, Fecci PE, Sampson JH, Dranoff G. An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev Vaccines 2013; 12:597-615. [PMID: 23750791 DOI: 10.1586/erv.13.41] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Outcome for glioblastoma (GBM), the most common primary CNS malignancy, remains poor. The overall survival benefit recently achieved with immunotherapeutics for melanoma and prostate cancer support evaluation of immunotherapies for other challenging cancers, including GBM. Much historical dogma depicting the CNS as immunoprivileged has been replaced by data demonstrating CNS immunocompetence and active interaction with the peripheral immune system. Several glioma antigens have been identified for potential immunotherapeutic exploitation. Active immunotherapy studies for GBM, supported by preclinical data, have focused on tumor lysate and synthetic antigen vaccination strategies. Results to date confirm consistent safety, including a lack of autoimmune reactivity; however, modest efficacy and variable immunogenicity have been observed. These findings underscore the need to optimize vaccination variables and to address challenges posed by systemic and local immunosuppression inherent to GBM tumors. Additional immunotherapy strategies are also in development for GBM. Future studies may consider combinatorial immunotherapy strategies with complimentary actions.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
410
|
Quan M, Wang P, Cui J, Gao Y, Xie K. The roles of FOXM1 in pancreatic stem cells and carcinogenesis. Mol Cancer 2013; 12:159. [PMID: 24325450 PMCID: PMC3924162 DOI: 10.1186/1476-4598-12-159] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 11/28/2013] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the poorest prognoses among all cancers. Over the past several decades, investigators have made great advances in the research of PDAC pathogenesis. Importantly, identification of pancreatic cancer stem cells (PCSCs) in pancreatic cancer cases has increased our understanding of PDAC biology and therapy. PCSCs are responsible for pancreatic tumorigenesis and tumor progression via a number of mechanisms, including extensive proliferation, self-renewal, high tumorigenic ability, high propensity for invasiveness and metastasis, and resistance to conventional treatment. Furthermore, emerging evidence suggests that PCSCs are involved in the malignant transformation of pancreatic intraepithelial neoplasia. The molecular mechanisms that control PCSCs are related to alterations of various signaling pathways, for instance, Hedgehog, Notch, Wnt, B-cell-specific Moloney murine leukemia virus insertion site 1, phosphoinositide 3-kinase/AKT, and Nodal/Activin. Also, authors have reported that the proliferation-specific transcriptional factor Forkhead box protein M1 is involved in PCSC self-renewal and proliferation. In this review, we describe the current knowledge about the signaling pathways related to PCSCs and the early stages of PDAC development, highlighting the pivotal roles of Forkhead box protein M1 in PCSCs and their impacts on the development and progression of pancreatic intraepithelial neoplasia.
Collapse
Affiliation(s)
| | | | | | | | - Keping Xie
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
411
|
Sarkar S, Döring A, Zemp FJ, Silva C, Lun X, Wang X, Kelly J, Hader W, Hamilton M, Mercier P, Dunn JF, Kinniburgh D, van Rooijen N, Robbins S, Forsyth P, Cairncross G, Weiss S, Yong VW. Therapeutic activation of macrophages and microglia to suppress brain tumor-initiating cells. Nat Neurosci 2013; 17:46-55. [PMID: 24316889 DOI: 10.1038/nn.3597] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/06/2013] [Indexed: 12/15/2022]
Abstract
Brain tumor initiating cells (BTICs) contribute to the genesis and recurrence of gliomas. We examined whether the microglia and macrophages that are abundant in gliomas alter BTIC growth. We found that microglia derived from non-glioma human subjects markedly mitigated the sphere-forming capacity of glioma patient-derived BTICs in culture by inducing the expression of genes that control cell cycle arrest and differentiation. This sphere-reducing effect was mimicked by macrophages, but not by neurons or astrocytes. Using a drug screen, we validated amphotericin B (AmpB) as an activator of monocytoid cells and found that AmpB enhanced the microglial reduction of BTIC spheres. In mice harboring intracranial mouse or patient-derived BTICs, daily systemic treatment with non-toxic doses of AmpB substantially prolonged life. Notably, microglia and monocytes cultured from glioma patients were inefficient at reducing the sphere-forming capacity of autologous BTICs, but this was rectified by AmpB. These results provide new insights into the treatment of gliomas.
Collapse
Affiliation(s)
- Susobhan Sarkar
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Axinia Döring
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. [3]
| | - Franz J Zemp
- 1] The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada. [2]
| | - Claudia Silva
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Xueqing Lun
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Xiuling Wang
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - John Kelly
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Walter Hader
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mark Hamilton
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Philippe Mercier
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F Dunn
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dave Kinniburgh
- Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Stephen Robbins
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Peter Forsyth
- The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Gregory Cairncross
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] The Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- 1] Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. [2] Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
412
|
Lu ZH, Lv K, Zhang JS, Dai CG, Liu B, Ma XY, He LM, Jia JY, Chen YM, Dai XL, Wang AD, Dong J, Zhang QB, Lan Q, Huang Q. Establishment of a green fluorescent protein tracing murine model focused on the functions of host components in necrosis repair and the niche of subcutaneously implanted glioma. Oncol Rep 2013; 31:657-64. [PMID: 24284913 DOI: 10.3892/or.2013.2873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/13/2013] [Indexed: 11/05/2022] Open
Abstract
Due to progress in the research of glioma stem cells and the glioma niche, development of an animal model that facilitates the elucidation of the roles of the host tissue and cells is necessary. The aim of the present study was to develop a subcutaneous xenograft green fluorescent protein nude mouse model and use this model to analyze the roles of host cells in tumor necrosis repair. Tumors derived from the human glioma stem/progenitor cell line SU3 were subcutaneously implanted in green fluorescent protein nude mice. The implanted tumors were then passed from animal to animal for 10 generations. Finally, subcutaneous xenografts were assayed with traditional pathology, immunopathological techniques and fluorescence photography. For each generation, the tumorigenicity rate was 100%. Subcutaneous xenografts were rich in blood vessels, and necrotic and hemorrhagic foci, which highly expressed hypoxia-inducible factor-1α, tumor necrosis factor, Ki-67, CD68 and CD11b. In the interstitial tissue, particularly in old hemorrhagic foci, there were numerous cells expressing green fluorescent protein, CD68 and CD11b. Green fluorescent protein nude mouse subcutaneous xenografts not only consistently maintained the high invasiveness and tumorigenicity of glioma stem/progenitor cells, but also consisted of a high concentration of tumor blood vessels and necrotic and hemorrhagic foci. Subcutaneous xenografts also expressed high levels of tumor microenvironment-related proteins and host-derived tumor interstitial molecules. The model has significant potential for further research on tumor tissue remodeling and the tumor microenvironment.
Collapse
Affiliation(s)
- Zhao-Hui Lu
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Ke Lv
- Neurosurgical Department of Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Jin-Shi Zhang
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Chun-Gang Dai
- Neurosurgical Department of Suzhou Seventh People's Hospital, Suzhou 215004, P.R. China
| | - Bin Liu
- Neurosurgical Department of Suzhou Seventh People's Hospital, Suzhou 215004, P.R. China
| | - Xiao-Yu Ma
- Neurosurgical Department of Suzhou Seventh People's Hospital, Suzhou 215004, P.R. China
| | - Lin-Ming He
- Neurosurgical Department of Suzhou Seventh People's Hospital, Suzhou 215004, P.R. China
| | - Jing-Yun Jia
- Neurosurgical Department of Suzhou Seventh People's Hospital, Suzhou 215004, P.R. China
| | - Yan-Ming Chen
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Xing-Liang Dai
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Ai-Dong Wang
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Jun Dong
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Quan-Bin Zhang
- Neurosurgical Department of Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Qing Lan
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| | - Qiang Huang
- Neurosurgical Department of The Second Affiliated Hospital, Soochow University, Suzhou 215004, P.R. China
| |
Collapse
|
413
|
Skoda J, Neradil J, Zitterbart K, Sterba J, Veselska R. EGFR signaling in the HGG-02 glioblastoma cell line with an unusual loss of EGFR gene copy. Oncol Rep 2013; 31:480-7. [PMID: 24270553 DOI: 10.3892/or.2013.2864] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/24/2013] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) gene amplification and the overexpression of EGFR are described as common features of glioblastoma multiforme (GBM). Nevertheless, we previously reported the loss of EGFR gene copy in a GBM specimen from a patient with an unusually favorable course of the disease, and the HGG-02 cell line with this aberration was successfully derived from this tumor. Here, we present a detailed analysis of changes in gene expression and cell signaling in the HGG-02 cell line; the GM7 reference cell line with a standard EGFR gene copy number derived from a very aggressive GBM was used as a control. We confirmed the downregulation of EGFR expression and signaling in HGG-02 cells using different methods (RTK analysis, gene profiling and RT-PCR). Other changes that may have contributed to the non-aggressive phenotype of the primary tumor were identified, including the downregulated phosphorylation of the Axl and Trk receptors, as well as increased activity of JNK and p38 kinases. Notably, differences in PDGF signaling were detected in both of these cell lines; HGG-02 cells preferentially expressed and signaled through PDGFRα, and PDGFRβ was strongly overexpressed and phosphorylated in the GM7 reference cell line. Using expression profiling of cancer-related genes, we revealed the specific profile of HGG-02 cells that included upregulated tumor-suppressors as well as downregulated genes associated with the extracellular matrix. This study represents the first comprehensive analysis of gene expression and cell signaling in glioblastoma cells with lower EGFR gene dosage. As indicated by our results, the TAM receptors, Trk receptors and PDGFRs need to be investigated further since their regulation appears to be important for glioblastoma biological features as well as the clinical course of the disease.
Collapse
Affiliation(s)
- Jan Skoda
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | | | | | | |
Collapse
|
414
|
NF-κB-induced IL-6 ensures STAT3 activation and tumor aggressiveness in glioblastoma. PLoS One 2013; 8:e78728. [PMID: 24244348 PMCID: PMC3823708 DOI: 10.1371/journal.pone.0078728] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/16/2013] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive, neurologically destructive and deadly tumor of the central nervous system (CNS). In GBM, the transcription factors NF-κB and STAT3 are aberrantly activated and associated with tumor cell proliferation, survival, invasion and chemoresistance. In addition, common activators of NF-κB and STAT3, including TNF-α and IL-6, respectively, are abundantly expressed in GBM tumors. Herein, we sought to elucidate the signaling crosstalk that occurs between the NF-κB and STAT3 pathways in GBM tumors. Using cultured GBM cell lines as well as primary human GBM xenografts, we elucidated the signaling crosstalk between the NF-κB and STAT3 pathways utilizing approaches that either a) reduce NF-κB p65 expression, b) inhibit NF-κB activation, c) interfere with IL-6 signaling, or d) inhibit STAT3 activation. Using the clinically relevant human GBM xenograft model, we assessed the efficacy of inhibiting NF-κB and/or STAT3 alone or in combination in mice bearing intracranial xenograft tumors in vivo. We demonstrate that TNF-α-induced activation of NF-κB is sufficient to induce IL-6 expression, activate STAT3, and elevate STAT3 target gene expression in GBM cell lines and human GBM xenografts in vitro. Moreover, the combined inhibition of NF-κB and STAT3 signaling significantly increases survival of mice bearing intracranial tumors. We propose that in GBM, the activation of NF-κB ensures subsequent STAT3 activation through the expression of IL-6. These data verify that pharmacological interventions to effectively inhibit the activity of both NF-κB and STAT3 transcription factors must be used in order to reduce glioma size and aggressiveness.
Collapse
|
415
|
Bielamowicz K, Khawja S, Ahmed N. Adoptive cell therapies for glioblastoma. Front Oncol 2013; 3:275. [PMID: 24273748 PMCID: PMC3823029 DOI: 10.3389/fonc.2013.00275] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/25/2013] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is the most common and most aggressive primary brain malignancy and, as it stands, is virtually incurable. With the current standard of care, maximum feasible surgical resection followed by radical radiotherapy and adjuvant temozolomide, survival rates are at a median of 14.6 months from diagnosis in molecularly unselected patients (1). Collectively, the current knowledge suggests that the continued tumor growth and survival is in part due to failure to mount an effective immune response. While this tolerance is subtended by the tumor being utterly “self,” it is to a great extent due to local and systemic immune compromise mediated by the tumor. Different cell modalities including lymphokine-activated killer cells, natural killer cells, cytotoxic T lymphocytes, and transgenic chimeric antigen receptor or αβ T cell receptor grafted T cells are being explored to recover and or redirect the specificity of the cellular arm of the immune system toward the tumor complex. Promising phase I/II trials of such modalities have shown early indications of potential efficacy while maintaining a favorable toxicity profile. Efficacy will need to be formally tested in phase II/III clinical trials. Given the high morbidity and mortality of GBM, it is imperative to further investigate and possibly integrate such novel cell-based therapies into the current standards-of-care and herein we collectively assess and critique the state-of-the-knowledge pertaining to these efforts.
Collapse
Affiliation(s)
- Kevin Bielamowicz
- Center for Cell and Gene Therapy, Baylor College of Medicine , Houston, TX , USA ; Texas Children's Cancer Center, Baylor College of Medicine , Houston, TX , USA ; Department of Pediatrics, Baylor College of Medicine , Houston, TX , USA ; Baylor College of Medicine , Houston, TX , USA
| | | | | |
Collapse
|
416
|
Wang XF, Wang HS, Zhang F, Guo Q, Wang H, Wang KF, Zhang G, Bu XZ, Cai SH, Du J. Nodal promotes the generation of M2-like macrophages and downregulates the expression of IL-12. Eur J Immunol 2013; 44:173-83. [DOI: 10.1002/eji.201343535] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/06/2013] [Accepted: 09/24/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Xian-Feng Wang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Fan Zhang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Qiang Guo
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Hao Wang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Ke-Fang Wang
- Department of Obstetrics and Gynecology; Beijing Anzhen Hospital, Capital Medical University; Beijing P.R. China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Xian-zhang Bu
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| | - Shao-Hui Cai
- Department of Pharmacology; School of Pharmaceutical Sciences, Jinan University; Guangzhou P.R. China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy; School of Pharmaceutical Sciences, Sun Yat-sen University; Guangzhou P.R. China
| |
Collapse
|
417
|
Coniglio SJ, Segall JE. Review: molecular mechanism of microglia stimulated glioblastoma invasion. Matrix Biol 2013; 32:372-80. [PMID: 23933178 DOI: 10.1016/j.matbio.2013.07.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 07/28/2013] [Accepted: 07/28/2013] [Indexed: 01/01/2023]
Abstract
Glioblastoma multiforme is one of the deadliest human cancers and is characterized by a high degree of microglia and macrophage infiltration. The role of these glioma infiltrating macrophages (GIMs) in disease progression has been the subject of recent investigation. While initially thought to reflect an immune response to the tumor, the balance of evidence clearly suggests GIMs can have potent tumor-tropic functions and assist in glioma cell growth and infiltration into normal brain. In this review, we focus on the evidence for GIMs aiding mediating glioblastoma motility and invasion. We survey the literature for molecular pathways that are involved in paracrine interaction between glioma cells and GIMs and assess which of these might serve as attractive targets for therapeutic intervention.
Collapse
Affiliation(s)
- Salvatore J Coniglio
- Albert Einstein College of Medicine, Department of Anatomy and Structural Biology, Bronx, NY 10461, United States.
| | | |
Collapse
|
418
|
Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, Olson OC, Quick ML, Huse JT, Teijeiro V, Setty M, Leslie CS, Oei Y, Pedraza A, Zhang J, Brennan CW, Sutton JC, Holland EC, Daniel D, Joyce JA. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med 2013; 19:1264-72. [PMID: 24056773 PMCID: PMC3840724 DOI: 10.1038/nm.3337] [Citation(s) in RCA: 1706] [Impact Index Per Article: 155.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 08/14/2013] [Indexed: 11/26/2022]
Abstract
Glioblastoma multiforme (GBM) comprises several molecular subtypes including proneural GBM. Most therapeutic approaches targeting glioma cells have failed. An alternative strategy is to target cells in the glioma microenvironment, such as tumor-associated macrophages and microglia (TAMs). Macrophages depend upon colony stimulating factor (CSF)-1 for differentiation and survival. A CSF-1R inhibitor was used to target TAMs in a mouse proneural GBM model, which dramatically increased survival, and regressed established tumors. CSF-1R blockade additionally slowed intracranial growth of patient-derived glioma xenografts. Surprisingly, TAMs were not depleted in treated mice. Instead, glioma-secreted factors including GM-CSF and IFN-γ facilitated TAM survival in the context of CSF-1R inhibition. Alternatively activated/ M2 macrophage markers decreased in surviving TAMs, consistent with impaired tumor-promoting functions. These gene signatures were associated with enhanced survival in proneural GBM patients. Our results identify TAMs as a promising therapeutic target for proneural gliomas, and establish the translational potential of CSF-1R inhibition for GBM.
Collapse
Affiliation(s)
- Stephanie M Pyonteck
- 1] Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York, USA. [2]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
419
|
Ochs K, Sahm F, Opitz CA, Lanz TV, Oezen I, Couraud PO, von Deimling A, Wick W, Platten M. Immature mesenchymal stem cell-like pericytes as mediators of immunosuppression in human malignant glioma. J Neuroimmunol 2013; 265:106-16. [PMID: 24090655 DOI: 10.1016/j.jneuroim.2013.09.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 09/11/2013] [Accepted: 09/13/2013] [Indexed: 02/08/2023]
Abstract
Malignant gliomas are primary brain tumors characterized by profound local immunosuppression. While the remarkable plasticity of perivascular cells - resembling mesenchymal stem cells (MSC) - in malignant gliomas and their contribution to angiogenesis is increasingly recognized, their role as potential mediators of immunosuppression is unknown. Here we demonstrate that FACS-sorted malignant glioma-derived pericytes (HMGP) were characterized by the expression of CD90, CD248, and platelet-derived growth factor receptor-β (PDGFR-β). HMGP shared this expression profile with human brain vascular pericytes (HBVP) and human MSC (HMSC) but not human cerebral microvascular endothelial cells (HCMEC). CD90+PDGFR-β+perivascular cells distinct from CD31+ endothelial cells accumulated in human gliomas with increasing degree of malignancy and negatively correlated with the presence of blood vessel-associated leukocytes and CD8+ T cells. Cultured CD90+PDGFR-β+HBVP were equally capable of suppressing allogeneic or mitogen-activated T cell responses as human MSC. HMGP, HBVP and HMSC expressed prostaglandin E synthase (PGES), inducible nitric oxide synthase (iNOS), human leukocyte antigen-G (HLA-G), hepatocyte growth factor (HGF) and transforming growth factor-β (TGF-β). These factors but not indoleamine 2,3-dioxygenase-mediated conversion of tryptophan to kynurenine functionally contributed to immunosuppression of immature pericytes. Our data provide evidence that human cerebral CD90+ perivascular cells possess T cell inhibitory capability comparable to human MSC and suggest that these cells, besides their critical role in tumor vascularization, also promote local immunosuppression in malignant gliomas and possibly other brain diseases.
Collapse
Affiliation(s)
- Katharina Ochs
- Department of Neurooncology, University Hospital Heidelberg and National Center for Tumor Diseases, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
420
|
Bhat KP, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, Wani K, Heathcock L, James JD, Goodman LD, Conroy S, Long L, Lelic N, Wang S, Gumin J, Raj D, Kodama Y, Raghunathan A, Olar A, Joshi K, Pelloski CE, Heimberger A, Kim SH, Cahill DP, Rao G, Den Dunnen WF, Boddeke HW, Phillips HS, Nakano I, Lang FF, Colman H, Sulman EP, Aldape K. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell 2013; 24:331-46. [PMID: 23993863 PMCID: PMC3817560 DOI: 10.1016/j.ccr.2013.08.001] [Citation(s) in RCA: 798] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 06/24/2013] [Accepted: 08/01/2013] [Indexed: 01/08/2023]
Abstract
Despite extensive study, few therapeutic targets have been identified for glioblastoma (GBM). Here we show that patient-derived glioma sphere cultures (GSCs) that resemble either the proneural (PN) or mesenchymal (MES) transcriptomal subtypes differ significantly in their biological characteristics. Moreover, we found that a subset of the PN GSCs undergoes differentiation to a MES state in a TNF-α/NF-κB-dependent manner with an associated enrichment of CD44 subpopulations and radioresistant phenotypes. We present data to suggest that the tumor microenvironment cell types such as macrophages/microglia may play an integral role in this process. We further show that the MES signature, CD44 expression, and NF-κB activation correlate with poor radiation response and shorter survival in patients with GBM.
Collapse
Affiliation(s)
- Krishna P.L. Bhat
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: ; ;
| | - Veerakumar Balasubramaniyan
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | | | - Ravesanker Ezhilarasan
- Department of Radiation Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Karlijn Hummelink
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Faith Hollingsworth
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Khalida Wani
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Lindsey Heathcock
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Johanna D. James
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Lindsey D. Goodman
- Department of Radiation Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Siobhan Conroy
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Lihong Long
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Nina Lelic
- Deparment of Neurosurgery, Massachusetts General Hospital/Brain Tumor Center, Boston, MA 02114, USA
| | - Suzhen Wang
- Department of Neuro-oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Divya Raj
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Yoshinori Kodama
- Division of Pathology, Osaka National Hospital, National Hospital Organization, Chuo-ku, Osaka 540-0006, Japan
| | | | - Adriana Olar
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Kaushal Joshi
- Department of Neurosurgery, The Ohio State University, Columbus, OH 43210, USA
| | | | - Amy Heimberger
- Department of Neurosurgery, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, 120-752, Korea
| | - Daniel P. Cahill
- Deparment of Neurosurgery, Massachusetts General Hospital/Brain Tumor Center, Boston, MA 02114, USA
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Wilfred F.A. Den Dunnen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Hendrikus W.G.M. Boddeke
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Heidi S. Phillips
- Department of Tumor Biology and Angiogenesis, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ichiro Nakano
- Department of Neurosurgery, The Ohio State University, Columbus, OH 43210, USA
| | - Frederick F. Lang
- Department of Neurosurgery, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Howard Colman
- Department of Neurosurgery, and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84132, USA
| | - Erik P. Sulman
- Department of Radiation Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: ; ;
| | - Kenneth Aldape
- Department of Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: ; ;
| |
Collapse
|
421
|
A cytokine cocktail directly modulates the phenotype of DC-enriched anti-tumor T cells to convey potent anti-tumor activities in a murine model. Cancer Immunol Immunother 2013; 62:1649-62. [PMID: 23982483 DOI: 10.1007/s00262-013-1464-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/07/2013] [Indexed: 10/26/2022]
Abstract
Adoptive cell transfer (ACT) using ex vivo-expanded anti-tumor T cells such as tumor-infiltrated lymphocytes or genetically engineered T cells potently eradicates established tumors. However, these two approaches possess obvious limitations. Therefore, we established a novel methodology using total tumor RNA (ttRNA) to prime dendritic cells (DC) as a platform for the ex vivo generation of anti-tumor T cells. We evaluated the antigen-specific expansion and recognition of T cells generated by the ttRNA-DC-T platform, and directly modulated the differentiation status of these ex vivo-expanded T cells with a cytokine cocktail. Furthermore, we evaluated the persistence and in vivo anti-tumor efficacy of these T cells through murine xenograft and syngeneic tumor models. During ex vivo culture, IL-2 preferentially expanded CD4 subset, while IL-7 enabled homeostatic proliferation from the original precursors. T cells tended to lose CD62L during ex vivo culture using IL-2; however, IL-12 could maintain high levels of CD62L by increasing expression on effector T cells (Tem). In addition, we validated that OVA RNA-DC only selectively expanded T cells in an antigen-specific manner. A cytokine cocktail excluding the use of IL-2 greatly increased CD62Lhigh T cells which specifically recognized tumor cells, engrafted better in a xenograft model and exhibited superior anti-tumor activities in a syngeneic intracranial model. ACT using the ex vivo ttRNA-DC-T platform in conjunction with a cytokine cocktail generated potent CD62Lhigh anti-tumor T cells and imposes a novel T cell-based therapeutic with the potential to treat brain tumors and other cancers.
Collapse
|
422
|
D'Alessandro G, Catalano M, Sciaccaluga M, Chece G, Cipriani R, Rosito M, Grimaldi A, Lauro C, Cantore G, Santoro A, Fioretti B, Franciolini F, Wulff H, Limatola C. KCa3.1 channels are involved in the infiltrative behavior of glioblastoma in vivo. Cell Death Dis 2013; 4:e773. [PMID: 23949222 PMCID: PMC3763441 DOI: 10.1038/cddis.2013.279] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/25/2013] [Accepted: 07/02/2013] [Indexed: 01/03/2023]
Abstract
Glioblastoma multiforme (GBM) is a diffuse brain tumor characterized by high infiltration in the brain parenchyma rendering the tumor difficult to eradicate by neurosurgery. Efforts to identify molecular targets involved in the invasive behavior of GBM suggested ion channel inhibition as a promising therapeutic approach. To determine if the Ca(2+)-dependent K(+) channel KCa3.1 could represent a key element for GBM brain infiltration, human GL-15 cells were xenografted into the brain of SCID mice that were then treated with the specific KCa3.1 blocker TRAM-34 (1-((2-chlorophenyl) (diphenyl)methyl)-1H-pyrazole). After 5 weeks of treatment, immunofluorescence analyses of cerebral slices revealed reduced tumor infiltration and astrogliosis surrounding the tumor, compared with untreated mice. Significant reduction of tumor infiltration was also observed in the brain of mice transplanted with KCa3.1-silenced GL-15 cells, indicating a direct effect of TRAM-34 on GBM-expressed KCa3.1 channels. As KCa3.1 channels are also expressed on microglia, we investigated the effects of TRAM-34 on microglia activation in GL-15 transplanted mice and found a reduction of CD68 staining in treated mice. Similar results were observed in vitro where TRAM-34 reduced both phagocytosis and chemotactic activity of primary microglia exposed to GBM-conditioned medium. Taken together, these results indicate that KCa3.1 activity has an important role in GBM invasiveness in vivo and that its inhibition directly affects glioma cell migration and reduces astrocytosis and microglia activation in response to tumor-released factors. KCa3.1 channel inhibition therefore constitutes a potential novel therapeutic approach to reduce GBM spreading into the surrounding tissue.
Collapse
Affiliation(s)
- G D'Alessandro
- Institute Pasteur, Cenci Bolognetti Foundation, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
423
|
The controversial role of microglia in malignant gliomas. Clin Dev Immunol 2013; 2013:285246. [PMID: 23983766 PMCID: PMC3741958 DOI: 10.1155/2013/285246] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/19/2013] [Indexed: 01/01/2023]
Abstract
Malignant gliomas contain stroma and a variety of immune cells including abundant activated microglia/macrophages. Mounting evidence indicates that the glioma microenvironment converts the glioma-associated microglia/macrophages (GAMs) into glioma-supportive, immunosuppressive cells; however, GAMs can retain intrinsic anti-tumor properties. Here, we review and discuss this duality and the potential therapeutic strategies that may inhibit their glioma-supportive and propagating functions.
Collapse
|
424
|
Brain tumor stem cells: Molecular characteristics and their impact on therapy. Mol Aspects Med 2013; 39:82-101. [PMID: 23831316 DOI: 10.1016/j.mam.2013.06.004] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 06/14/2013] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM) is the most prevalent primary brain tumor and ranks among the most lethal of human cancers with conventional therapy offering only palliation. Great strides have been made in understanding brain cancer genetics and modeling these tumors with new targeted therapies being tested, but these advances have not translated into substantially improved patient outcomes. Multiple chemotherapeutic agents, including temozolomide, the first-line treatment for glioblastoma, have been developed to kill cancer cells. However, the response to temozolomide in GBM is modest. Radiation is also moderately effective but this approach is plagued by limitations due to collateral radiation damage to healthy brain tissue and development of radioresistance. Therapeutic resistance is attributed at least in part to a cell population within the tumor that possesses stem-like characteristics and tumor propagating capabilities, referred to as cancer stem cells. Within GBM, the intratumoral heterogeneity is derived from a combination of regional genetic variance and a cellular hierarchy often regulated by distinct cancer stem cell niches, most notably perivascular and hypoxic regions. With the recent emergence as a key player in tumor biology, cancer stem cells have symbiotic relationships with the tumor microenvironment, oncogenic signaling pathways, and epigenetic modifications. The origins of cancer stem cells and their contributions to brain tumor growth and therapeutic resistance are under active investigation with novel anti-cancer stem cell therapies offering potential new hope for this lethal disease.
Collapse
|
425
|
Ellert-Miklaszewska A, Dabrowski M, Lipko M, Sliwa M, Maleszewska M, Kaminska B. Molecular definition of the pro-tumorigenic phenotype of glioma-activated microglia. Glia 2013; 61:1178-90. [PMID: 23650109 DOI: 10.1002/glia.22510] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 03/20/2013] [Indexed: 12/16/2023]
Abstract
Microglia are myeloid cells residing in the central nervous system that participate in inflammatory responses and could promote injury and repair. Gliomas attract microglia and polarize them into tumor-supporting cells that participate in matrix remodeling, invasion, angiogenesis, and suppression of adaptive immunity. Although signaling pathways and critical regulators underlying classical inflammation are well established, signal transduction and transcriptional circuits underlying the alternative activation of microglia are poorly known. Using primary rat microglial cultures exposed to glioma conditioned medium or lipopolysaccharide (LPS), we demonstrate that microglia adapt different fates and polarize into pro-inflammatory or alternatively activated cells. Glioma-derived factors increased cell motility, phagocytosis, and sustained proliferation of microglial cells that was mediated by enhanced focal adhesion kinase and PI-3K/Akt signaling. The signals from glioma cells induced ERK and p38 MAPK but not JNK signaling and failed to activate pro-inflammatory Stat1 and NFκB signaling in microglial cells. Transcriptome analysis of microglial cultures at 6 h after exposure to glioma-conditioned medium or LPS revealed different patterns of gene expression. Glioma-induced activation was associated with induction of genes coding for ID (inhibitor of DNA binding) 1/3 and c-Myc, markers of the alternative phenotype Arg1, MT1-MMP, CXCL14, and numerous cytokines/chemokines implicated in immune cell trafficking. Many classical inflammation-related genes and signaling pathways failed to be induced. Our study indicates for the first time molecular pathways that direct microglia toward the pro-invasive, immunosuppressive phenotype.
Collapse
Affiliation(s)
- Aleksandra Ellert-Miklaszewska
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 3 Pasteur str., Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
426
|
Microglia and macrophages in malignant gliomas: recent discoveries and implications for promising therapies. Clin Dev Immunol 2013; 2013:264124. [PMID: 23864876 PMCID: PMC3707269 DOI: 10.1155/2013/264124] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 06/03/2013] [Indexed: 01/05/2023]
Abstract
Malignant gliomas are the most common primary brain tumors. Their deadliest manifestation, glioblastoma multiforme (GBM), accounts for 15% of all primary brain tumors and is associated with a median survival of only 15 months even after multimodal therapy. There is substantial presence of microglia and macrophages within and surrounding brain tumors. These immune cells acquire an alternatively activated phenotype with potent tumor-tropic functions that contribute to glioma growth and invasion. In this review, we briefly summarize recent data that has been reported on the interaction of microglia/macrophages with brain tumors and discuss potential application of these findings to the development of future antiglioma therapies.
Collapse
|
427
|
Multifaceted oncolytic virus therapy for glioblastoma in an immunocompetent cancer stem cell model. Proc Natl Acad Sci U S A 2013; 110:12006-11. [PMID: 23754388 DOI: 10.1073/pnas.1307935110] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma (World Health Organization grade IV) is an aggressive adult brain tumor that is inevitably fatal despite surgery, radiation, and chemotherapy. Treatment failures are attributed to combinations of cellular heterogeneity, including a subpopulation of often-resistant cancer stem cells, aberrant vasculature, and noteworthy immune suppression. Current preclinical models and treatment strategies do not incorporate or address all these features satisfactorily. Herein, we describe a murine glioblastoma stem cell (GSC) model that recapitulates tumor heterogeneity, invasiveness, vascularity, and immunosuppressive microenvironment in syngeneic immunocompetent mice and should prove useful for a range of therapeutic studies. Using this model, we tested a genetically engineered oncolytic herpes simplex virus that is armed with an immunomodulatory cytokine, interleukin 12 (G47-mIL12). G47Δ-mIL12 infects and replicates similarly to its unarmed oncolytic herpes simplex virus counterpart in mouse 005 GSCs in vitro, whereas in vivo, it significantly enhances survival in syngeneic mice bearing intracerebral 005 tumors. Mechanistically, G47-mIL12 targets not only GSCs but also increases IFN-γ release, inhibits angiogenesis, and reduces the number of regulatory T cells in the tumor. The increased efficacy is dependent upon T cells, but not natural killer cells. Taken together, our findings demonstrate that G47Δ-mIL12 provides a multifaceted approach to targeting GSCs, tumor microenvironment, and the immune system, with resultant therapeutic benefit in a stringent glioblastoma model.
Collapse
|
428
|
Sielska M, Przanowski P, Wylot B, Gabrusiewicz K, Maleszewska M, Kijewska M, Zawadzka M, Kucharska J, Vinnakota K, Kettenmann H, Kotulska K, Grajkowska W, Kaminska B. Distinct roles of CSF family cytokines in macrophage infiltration and activation in glioma progression and injury response. J Pathol 2013; 230:310-21. [DOI: 10.1002/path.4192] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 03/03/2013] [Accepted: 03/13/2013] [Indexed: 01/10/2023]
Affiliation(s)
- Malgorzata Sielska
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Piotr Przanowski
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Bartosz Wylot
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Konrad Gabrusiewicz
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Marta Maleszewska
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Magdalena Kijewska
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Malgorzata Zawadzka
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Joanna Kucharska
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| | - Katyayni Vinnakota
- Max Delbrück Center for Molecular Medicine; Cellular Neuroscience; Berlin Germany
| | - Helmut Kettenmann
- Max Delbrück Center for Molecular Medicine; Cellular Neuroscience; Berlin Germany
| | | | | | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center; Nencki Institute of Experimental Biology; Warsaw Poland
| |
Collapse
|
429
|
Heimberger AB, Gilbert M, Rao G, Wei J. MicroRNAs as novel immunotherapeutics. Oncoimmunology 2013; 2:e25124. [PMID: 24083077 PMCID: PMC3782519 DOI: 10.4161/onci.25124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 11/19/2022] Open
Abstract
A large unmet need exists for cost-effective, widely available antineoplastic immunotherapeutic agents with a robust translational potential. MicroRNAs (miRNAs) that regulate tumor-mediated immunosuppression or immune checkpoints can induce robust therapeutic immune responses, indicating that miRNAs may ultimately become part of the portfolio of anticancer immunotherapeutics.
Collapse
Affiliation(s)
- Amy B Heimberger
- Department of Neurosurgery; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | | | | | | |
Collapse
|
430
|
Hoepner S, Loh JMS, Riccadonna C, Derouazi M, Maroun CY, Dietrich PY, Walker PR. Synergy between CD8 T cells and Th1 or Th2 polarised CD4 T cells for adoptive immunotherapy of brain tumours. PLoS One 2013; 8:e63933. [PMID: 23717511 PMCID: PMC3662716 DOI: 10.1371/journal.pone.0063933] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/10/2013] [Indexed: 01/05/2023] Open
Abstract
The feasibility of cancer immunotherapy mediated by T lymphocytes is now a clinical reality. Indeed, many tumour associated antigens have been identified for cytotoxic CD8 T cells, which are believed to be key mediators of tumour rejection. However, for aggressive malignancies in specialised anatomic sites such as the brain, a limiting factor is suboptimal tumour infiltration by CD8 T cells. Here we take advantage of recent advances in T cell biology to differentially polarise CD4 T cells in order to explore their capacity to enhance immunotherapy. We used an adoptive cell therapy approach to work with clonal T cell populations of defined specificity. Th1 CD4 T cells preferentially homed to and accumulated within intracranial tumours compared with Th2 CD4 T cells. Moreover, tumour-antigen specific Th1 CD4 T cells enhanced CD8 T cell recruitment and function within the brain tumour bed. Survival of mice bearing intracranial tumours was significantly prolonged when CD4 and CD8 T cells were co-transferred. These results should encourage further definition of tumour antigens recognised by CD4 T cells, and exploitation of both CD4 and CD8 T cell subsets to optimise T cell therapy of cancer.
Collapse
Affiliation(s)
- Sabine Hoepner
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Jacelyn M. S. Loh
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Cristina Riccadonna
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Madiha Derouazi
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Céline Yacoub Maroun
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Paul R. Walker
- Centre of Oncology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
431
|
Tumor-associated macrophages in glioma: friend or foe? JOURNAL OF ONCOLOGY 2013; 2013:486912. [PMID: 23737783 PMCID: PMC3664503 DOI: 10.1155/2013/486912] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/08/2013] [Indexed: 12/31/2022]
Abstract
Tumor-associated macrophages (TAMs) contribute substantially to the tumor mass of gliomas and have been shown to play a major role in the creation of a tumor microenvironment that promotes tumor progression. Shortcomings of attempts at antiglioma immunotherapy may result from a failure to adequately address these effects. Emerging evidence supports an independent categorization of glioma TAMs as alternatively activated M2-type macrophages, in contrast to classically activated proinflammatory M1-type macrophages. These M2-type macrophages exert glioma-supportive effects through reduced anti-tumor functions, increased expression of immunosuppressive mediators, and nonimmune tumor promotion through expression of trophic and invasion-facilitating substances. Much of our work has demonstrated these features of glioma TAMs, and together with the supporting literature will be reviewed here. Additionally, the dynamics of glioma cell-TAM interaction over the course of tumor development remain poorly understood; our efforts to elucidate glioma cell-TAM dynamics are summarized. Finally, the molecular pathways which underlie M2-type TAM polarization and gene expression similarly require further investigation, and may present the most potent targets for immunotherapeutic intervention. Highlighting recent evidence implicating the transcription factor STAT3 in immunosuppressive tumorigenic glioma TAMs, we advocate for gene array-based approaches to identify yet unappreciated expression regulators and effector molecules important to M2-type glioma TAMs polarization and function within the glioma tumor microenvironment.
Collapse
|
432
|
Bovenberg MSS, Degeling MH, Tannous BA. Cell-based immunotherapy against gliomas: from bench to bedside. Mol Ther 2013; 21:1297-305. [PMID: 23648695 DOI: 10.1038/mt.2013.80] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/17/2013] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) comprises 51% of all gliomas and is the most malignant form of brain tumors with a median survival of 18-21 months. Standard-of-care treatment includes maximal surgical resection of the tumor mass in combination with radiation and chemotherapy. However, as the poor survival rate indicates, these treatments have not been effective in preventing disease progression. Cellular immunotherapy is currently being explored as therapeutic approach to treat malignant brain tumors. In this review, we discuss advances in active, passive, and vaccine-based immunotherapeutic strategies for gliomas both at the bench and in the clinic.
Collapse
Affiliation(s)
- M Sarah S Bovenberg
- Department of Neurology, Experimental Therapeutics and Molecular Imaging Laboratory, Neuroscience Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | |
Collapse
|
433
|
Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, Yang Y, McEnery K, Jethwa K, Gjyshi O, Qiao W, Levine NB, Lang FF, Rao G, Fuller GN, Calin GA, Heimberger AB. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res 2013; 73:3913-26. [PMID: 23636127 DOI: 10.1158/0008-5472.can-12-4318] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
miRNAs (miR) have been shown to modulate critical gene transcripts involved in tumorigenesis, but their role in tumor-mediated immunosuppression is largely unknown. On the basis of miRNA gene expression in gliomas using tissue microarrays, in situ hybridization, and molecular modeling, miR-124 was identified as a lead candidate for modulating STAT3 signaling, a key pathway mediating immunosuppression in the tumor microenvironment. miR-124 is absent in all grades and pathologic types of gliomas. Upon upregulating miR-124 in glioma cancer stem cells (gCSC), the STAT3 pathway was inhibited, and miR-124 reversed gCSC-mediated immunosuppression of T-cell proliferation and induction of forkhead box P3 (Foxp3)(+) regulatory T cells (Treg). Treatment of T cells from immunosuppressed glioblastoma patients with miR-124 induced marked effector response including upregulation of interleukin (IL)-2, IFN-γ, and TNF-α. Both systemic administration of miR-124 or adoptive miR-124-transfected T-cell transfers exerted potent anti-glioma therapeutic effects in clonotypic and genetically engineered murine models of glioblastoma and enhanced effector responses in the local tumor microenvironment. These therapeutic effects were ablated in both CD4(+)- and CD8(+)-depleted mice and nude mouse systems, indicating that the therapeutic effect of miR-124 depends on the presence of a T-cell-mediated antitumor immune response. Our findings highlight the potential application of miR-124 as a novel immunotherapeutic agent for neoplasms and serve as a model for identifying miRNAs that can be exploited as immunotherapeutics.
Collapse
Affiliation(s)
- Jun Wei
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
434
|
Hitting Them Where They Live: Targeting the Glioblastoma Perivascular Stem Cell Niche. CURRENT PATHOBIOLOGY REPORTS 2013; 1:101-110. [PMID: 23766946 DOI: 10.1007/s40139-013-0012-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Glioblastoma growth potential and resistance to therapy is currently largely attributed to a subset of tumor cells with stem-like properties. If correct, this means that cure will not be possible without eradication of the stem cell fraction and abrogation of those mechanisms through which stem cell activity is induced and maintained. Glioblastoma stem cell functions appear to be non-cell autonomous and the consequence of tumor cell residence within specialized domains such as the perivascular stem cell niche. In this review we consider the multiple cellular constituents of the perivascular niche, the molecular mechanisms that support niche structure and function and the implications of the perivascular localization of stem cells for anti-angiogenic approaches to cure.
Collapse
|
435
|
Filatova A, Acker T, Garvalov BK. The cancer stem cell niche(s): The crosstalk between glioma stem cells and their microenvironment. Biochim Biophys Acta Gen Subj 2013; 1830:2496-508. [DOI: 10.1016/j.bbagen.2012.10.008] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 09/27/2012] [Accepted: 10/10/2012] [Indexed: 01/14/2023]
|
436
|
Sierra A, Abiega O, Shahraz A, Neumann H. Janus-faced microglia: beneficial and detrimental consequences of microglial phagocytosis. Front Cell Neurosci 2013. [PMID: 23386811 DOI: 10.3389/fncel.2013.00006/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Microglia are the resident brain macrophages and they have been traditionally studied as orchestrators of the brain inflammatory response during infections and disease. In addition, microglia has a more benign, less explored role as the brain professional phagocytes. Phagocytosis is a term coined from the Greek to describe the receptor-mediated engulfment and degradation of dead cells and microbes. In addition, microglia phagocytoses brain-specific cargo, such as axonal and myelin debris in spinal cord injury or multiple sclerosis, amyloid-β deposits in Alzheimer's disease, and supernumerary synapses in postnatal development. Common mechanisms of recognition, engulfment, and degradation of the different types of cargo are assumed, but very little is known about the shared and specific molecules involved in the phagocytosis of each target by microglia. More importantly, the functional consequences of microglial phagocytosis remain largely unexplored. Overall, phagocytosis is considered a beneficial phenomenon, since it eliminates dead cells and induces an anti-inflammatory response. However, phagocytosis can also activate the respiratory burst, which produces toxic reactive oxygen species (ROS). Phagocytosis has been traditionally studied in pathological conditions, leading to the assumption that microglia have to be activated in order to become efficient phagocytes. Recent data, however, has shown that unchallenged microglia phagocytose apoptotic cells during development and in adult neurogenic niches, suggesting an overlooked role in brain remodeling throughout the normal lifespan. The present review will summarize the current state of the literature regarding the role of microglial phagocytosis in maintaining tissue homeostasis in health as in disease.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro-Basque Center for Neuroscience Zamudio, Spain ; Department of Neuroscience, University of the Basque Country EHU/UPV Leioa, Spain ; Ikerbasque-Basque Foundation for Science Bilbao, Spain
| | | | | | | |
Collapse
|
437
|
Sierra A, Abiega O, Shahraz A, Neumann H. Janus-faced microglia: beneficial and detrimental consequences of microglial phagocytosis. Front Cell Neurosci 2013; 7:6. [PMID: 23386811 PMCID: PMC3558702 DOI: 10.3389/fncel.2013.00006] [Citation(s) in RCA: 394] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/09/2013] [Indexed: 02/04/2023] Open
Abstract
Microglia are the resident brain macrophages and they have been traditionally studied as orchestrators of the brain inflammatory response during infections and disease. In addition, microglia has a more benign, less explored role as the brain professional phagocytes. Phagocytosis is a term coined from the Greek to describe the receptor-mediated engulfment and degradation of dead cells and microbes. In addition, microglia phagocytoses brain-specific cargo, such as axonal and myelin debris in spinal cord injury or multiple sclerosis, amyloid-β deposits in Alzheimer's disease, and supernumerary synapses in postnatal development. Common mechanisms of recognition, engulfment, and degradation of the different types of cargo are assumed, but very little is known about the shared and specific molecules involved in the phagocytosis of each target by microglia. More importantly, the functional consequences of microglial phagocytosis remain largely unexplored. Overall, phagocytosis is considered a beneficial phenomenon, since it eliminates dead cells and induces an anti-inflammatory response. However, phagocytosis can also activate the respiratory burst, which produces toxic reactive oxygen species (ROS). Phagocytosis has been traditionally studied in pathological conditions, leading to the assumption that microglia have to be activated in order to become efficient phagocytes. Recent data, however, has shown that unchallenged microglia phagocytose apoptotic cells during development and in adult neurogenic niches, suggesting an overlooked role in brain remodeling throughout the normal lifespan. The present review will summarize the current state of the literature regarding the role of microglial phagocytosis in maintaining tissue homeostasis in health as in disease.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro-Basque Center for Neuroscience Zamudio, Spain ; Department of Neuroscience, University of the Basque Country EHU/UPV Leioa, Spain ; Ikerbasque-Basque Foundation for Science Bilbao, Spain
| | | | | | | |
Collapse
|
438
|
Blaylock RL. Immunoexcitatory mechanisms in glioma proliferation, invasion and occasional metastasis. Surg Neurol Int 2013; 4:15. [PMID: 23493580 PMCID: PMC3589840 DOI: 10.4103/2152-7806.106577] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/12/2012] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence of an interaction between inflammatory cytokines and glutamate receptors among a number of neurological diseases including traumatic brain injuries, neurodegenerative diseases and central nervous system (CNS) infections. A number of recent studies have now suggested a strong relation between inflammatory mechanisms and excitatory cascades and these may play a role in glioma invasiveness and proliferation. Chronic inflammation appears to be a major initiating mechanism in most human cancers, involving cell-signaling pathways, which are responsible for cell cycling, cancer cell migration, invasion, tumor aggressiveness, and angiogenesis. It is less well appreciated that glutamate receptors also play a significant role in both proliferation and especially glioma invasion. There is some evidence that sustained elevations in glutamate may play a role in initiating certain cancers and new studies demonstrate an interaction between inflammation and glutamate receptors that may enhance tumor invasion and metastasis by affecting a number of cell-signaling mechanisms. These mechanisms are discussed in this paper as well as novel treatment options for reducing immune-glutamate promotion of cancer growth and invasion.
Collapse
Affiliation(s)
- Russell L Blaylock
- Theoretical Neurosciences LLC, Visiting Professor of Biology, Department of Biology, Belhaven University, Jackson, MS 39157, USA
| |
Collapse
|
439
|
Mo LJ, Ye HX, Mao Y, Yao Y, Zhang JM. B7-H4 expression is elevated in human U251 glioma stem-like cells and is inducible in monocytes cultured with U251 stem-like cell conditioned medium. CHINESE JOURNAL OF CANCER 2013; 32:653-60. [PMID: 23327799 PMCID: PMC3870849 DOI: 10.5732/cjc.012.10228] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Previous studies indicated that B7-H4, the youngest B7 family, negatively regulates T cell-mediated immunity and is significantly overexpressed in many human tumors. Tumor stem cells are purported to play a role in tumor renewal and resistance to radiation and chemotherapy. However, the link between B7-H4 and tumor stem cells is unclear. In this study, we investigated B7-H4 expression in the medium of human glioma U251 cell cultures. Immunofluorescence results showed that U251 cells cultured in serum-free medium (supplemented with 2% B27, 20 ng/mL epidermal growth factor, 20 ng/mL basic fibroblast growth factor) maintained stem-like cell characteristics, including expression of stem cell marker CD133 and the neural progenitor cell markers nestin and SOX2. In contrast, U251 cells cultured in serum-containing medium highly expressed differentiation marker glial fibrillary acidic protein. Flow cytometry analysis showed serum-free medium-cultured U251 cells expressed higher intracellular B7-H4 than serum-containing medium-cultured U251 cells (24%–35% vs. 8%–11%, P < 0.001). Immunofluorescence in purified monocytes from normal human peripheral blood mononuclear cells revealed moderate expression of B7-H4 after stimulation with conditioned medium from U251 cells cultured in serum-containing medium. Moreover, conditioned medium from U251 stem-like cells had a significant stimulation effect on B7-H4 expression compared with serum-containing conditioned medium (P < 0.01). Negative costimulatory molecule B7-H4 was preferentially expressed in U251 stem-like cells, and conditioned medium from these cells more effectively induced monocytes to express B7-H4 than conditioned medium from U251 cells cultured in the presence of serum. Our results show that U251 stem-like cells may play a more crucial role in tumor immunoloregulation with high expression of B7-H4.
Collapse
Affiliation(s)
- Lian-Jie Mo
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P. R. China. ,
| | | | | | | | | |
Collapse
|
440
|
Abstract
The brain is in many ways an immunologically and pharmacologically privileged site. The blood-brain barrier (BBB) of the cerebrovascular endothelium and its participation in the complex structure of the neurovascular unit (NVU) restrict access of immune cells and immune mediators to the central nervous system (CNS). In pathologic conditions, very well-organized immunologic responses can develop within the CNS, raising important questions about the real nature and the intrinsic and extrinsic regulation of this immune privilege. We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors. The goals of this review are to outline key scientific advances and the status of the science central to both the neuroinflammation and CNS barriers fields, and highlight the opportunities and priorities in advancing brain barriers research in the context of the larger immunology and neuroscience disciplines. This review article was developed from reports presented at the 2011 Annual Blood-Brain Barrier Consortium Meeting.
Collapse
|
441
|
Abstract
The investigation and development of the cancer stem cell (CSC) model has received much focus during these years. CSC is characterized as a small fraction of cancer cells that have an indefinite ability for self-renewal and pluripotency and are responsible for initiating and sustaining of the bulk of cancer. So, whether current treatment strategies, most of which target the rapid division of cancer cells, could interfere with the slow-cycling CSCs is broadly questioned. Meanwhile, however, the new understanding of tumorigenesis has led to the development of new drug screening strategies. Both stem cells and mesenchymal stem cells have been vigorously used in pre-clinical studies of their anti-tumor potential, mainly due to their inherent tumoritropic migratory properties and their ability to carry anti-tumor transgenes. Here, based on the tumorigenic and tumoritropic characteristics of CSCs, we proposed two hypotheses exploring possible usage of CSCs as novel anti-tumor agents and potential sources for tissue regeneration. Further experimental validation of these hypotheses may unravel some new research topics.
Collapse
|
442
|
Cancer stem cell hypothesis: a brief summary and two proposals. Cytotechnology 2012; 65:505-12. [PMID: 23250634 DOI: 10.1007/s10616-012-9517-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 11/16/2012] [Indexed: 12/18/2022] Open
Abstract
The investigation and development of the cancer stem cell (CSC) model has received much focus during these years. CSC is characterized as a small fraction of cancer cells that have an indefinite ability for self-renewal and pluripotency and are responsible for initiating and sustaining of the bulk of cancer. So, whether current treatment strategies, most of which target the rapid division of cancer cells, could interfere with the slow-cycling CSCs is broadly questioned. Meanwhile, however, the new understanding of tumorigenesis has led to the development of new drug screening strategies. Both stem cells and mesenchymal stem cells have been vigorously used in pre-clinical studies of their anti-tumor potential, mainly due to their inherent tumoritropic migratory properties and their ability to carry anti-tumor transgenes. Here, based on the tumorigenic and tumoritropic characteristics of CSCs, we proposed two hypotheses exploring possible usage of CSCs as novel anti-tumor agents and potential sources for tissue regeneration. Further experimental validation of these hypotheses may unravel some new research topics.
Collapse
|
443
|
Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2012. [DOI: 10.1002/path.4133 or 1=1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Alberto Mantovani
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Department of Biotechnology and Translational Medicine; University of Milan; Italy
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR); Singapore
| | - Maria Rosaria Galdiero
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Division of Clinical Immunology and Allergy; University of Naples Federico II; Naples Italy
| | - Antonio Sica
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Department of Pharmaceutical Sciences; Università del Piemonte Orientale ‘Amedeo Avogadro’; Novara Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Department of Biotechnology and Translational Medicine; University of Milan; Italy
| |
Collapse
|
444
|
Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2012. [DOI: 10.1002/path.4133\] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Alberto Mantovani
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Department of Biotechnology and Translational Medicine; University of Milan; Italy
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR); Singapore
| | - Maria Rosaria Galdiero
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Division of Clinical Immunology and Allergy; University of Naples Federico II; Naples Italy
| | - Antonio Sica
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Department of Pharmaceutical Sciences; Università del Piemonte Orientale ‘Amedeo Avogadro’; Novara Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center; Via Manzoni 56 20089 Rozzano Milan Italy
- Department of Biotechnology and Translational Medicine; University of Milan; Italy
| |
Collapse
|
445
|
Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2012; 229:176-85. [PMID: 23096265 DOI: 10.1002/path.4133] [Citation(s) in RCA: 1692] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 10/04/2012] [Accepted: 10/05/2012] [Indexed: 12/12/2022]
Abstract
Mononuclear phagocyte plasticity includes the expression of functions related to the resolution of inflammation, tissue repair and remodelling, particularly when these cells are set in an M2 or an M2-like activation mode. Macrophages are credited with an essential role in remodelling during ontogenesis. In extraembryonic life, under homeostatic conditions, the macrophage trophic and remodelling functions are recapitulated in tissues such as bone, mammary gland, decidua and placenta. In pathology, macrophages are key components of tissue repair and remodelling that occur during wound healing, allergy, parasite infection and cancer. Interaction with cells bearing stem or progenitor cell properties is likely an important component of the role of macrophages in repair and remodelling. These properties of cells of the monocyte-macrophage lineage may represent a tool and a target for therapeutic exploitation.
Collapse
Affiliation(s)
- Alberto Mantovani
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| | | | | | | | | |
Collapse
|
446
|
Kushchayev SV, Kushchayeva YS, Wiener PC, Scheck AC, Badie B, Preul MC. Monocyte-derived cells of the brain and malignant gliomas: the double face of Janus. World Neurosurg 2012. [PMID: 23178919 DOI: 10.1016/j.wneu.2012.11.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Monocyte-derived cells of the brain (MDCB) are a diverse group of functional immune cells that are also highly abundant in gliomas. There is growing evidence that MDCB play essential roles in the pathogenesis of gliomas. The aim of this review was to collate and systematize contemporary knowledge about these cells as they relate to glioma progression and antiglioblastoma therapeutic modalities with a view toward improved effectiveness of therapy. METHODS We reviewed relevant studies to construct a summary of different MDCB subpopulations in steady state and in malignant gliomas and discuss their role in the development of malignant gliomas and potential future therapies. RESULTS Current studies suggest that MDCB subsets display different phenotypes and differentiation potentials depending on their milieu in the brain and exposure to tumoral influences. MDCB possess specific and unique functions, including those that are protumoral and those that are antitumoral. CONCLUSIONS Elucidating the role of mononuclear-derived cells associated with gliomas is crucial in designing novel immunotherapy strategies. Much progress is needed to characterize markers to identify cell subsets and their specific regulatory roles. Investigation of MDCB can be clinically relevant. Specific MDCB populations potentially can be used for glioma therapy as a target or as cell vehicles that might deliver cytotoxic substances or processes to the glioma microenvironment.
Collapse
Affiliation(s)
- Sergiy V Kushchayev
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Yevgeniya S Kushchayeva
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA; Department of Surgery, Medstar Washington Hospital Center, Washington, DC, USA
| | - Philip C Wiener
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Adrienne C Scheck
- Neuro-oncology Research Laboratory, Division of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Behnam Badie
- Division of Neurosurgery, Department of Surgery, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Mark C Preul
- Neurosurgery Research Laboratory, Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.
| |
Collapse
|
447
|
Salinomycin as a drug for targeting human cancer stem cells. J Biomed Biotechnol 2012; 2012:950658. [PMID: 23251084 PMCID: PMC3516046 DOI: 10.1155/2012/950658] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/04/2012] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation of tumor cells that possess self-renewal and tumor initiation capacity and the ability to give rise to the heterogenous lineages of malignant cells that comprise a tumor. CSCs possess multiple intrinsic mechanisms of resistance to chemotherapeutic drugs, novel tumor-targeted drugs, and radiation therapy, allowing them to survive standard cancer therapies and to initiate tumor recurrence and metastasis. Various molecular complexes and pathways that confer resistance and survival of CSCs, including expression of ATP-binding cassette (ABC) drug transporters, activation of the Wnt/β-catenin, Hedgehog, Notch and PI3K/Akt/mTOR signaling pathways, and acquisition of epithelial-mesenchymal transition (EMT), have been identified recently. Salinomycin, a polyether ionophore antibiotic isolated from Streptomyces albus, has been shown to kill CSCs in different types of human cancers, most likely by interfering with ABC drug transporters, the Wnt/β-catenin signaling pathway, and other CSC pathways. Promising results from preclinical trials in human xenograft mice and a few clinical pilote studies reveal that salinomycin is able to effectively eliminate CSCs and to induce partial clinical regression of heavily pretreated and therapy-resistant cancers. The ability of salinomycin to kill both CSCs and therapy-resistant cancer cells may define the compound as a novel and an effective anticancer drug.
Collapse
|
448
|
Abstract
OBJECTIVE Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant tumor of the central nervous system. Its pathogenesis remains unknown. Like glioblastomas, AT/RTs contain brain cancer stem cells (CSCs) that suppress the immunity of patients and are resistant to conventional chemotherapy and radiation therapy. Considerable infiltration of immune cells, including macrophages/microglia, dendritic cells and T-cells, has been noted in glioblastomas, which correlates with poor prognosis. The present study examines the significance of infiltrating immune cells in four cases of AT/RT; including one associated with an autoimmune disease, Henoch-Schonlein purpura. METHODS Tumor tissues from four patients with AT/RT were analyzed and compared with those from four patients with glioblastomas. The frequency of immune cells, including CD68+, CD4+, and CD8+ cells, was assessed by scoring for statistical analysis. RESULTS The infiltration of immune cells was identified in the case of AT/RT associated with HSP and three other cases of infratentorial AT/RTs. Moderate infiltration of CD68+ macrophages/microglia and CD4+ cells was noted in AT/RTs with no significant difference from that in glioblastomas (p > 0.05). However, the infiltration of CD8+ T-cells was significantly higher in AT/RTs than that in glioblastomas (p < 0.05); CD4+/CD8+ ratio was significantly lower in AT/RTs than that in glioblastomas (p < 0.05). In addition, eosinophils were found in all AT/RTs, but not in glioblastomas. CONCLUSIONS These findings suggest an immune microenvironment of AT/RTs with more immune effectors than glioblastomas. Our observation contributes to understanding the growth environment of AT/RTs for which adjuvant immunotherapy may be potentially beneficial.
Collapse
|
449
|
Komohara Y, Horlad H, Ohnishi K, Fujiwara Y, Bai B, Nakagawa T, Suzu S, Nakamura H, Kuratsu JI, Takeya M. Importance of direct macrophage-tumor cell interaction on progression of human glioma. Cancer Sci 2012; 103:2165-72. [PMID: 22957741 DOI: 10.1111/cas.12015] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/31/2012] [Accepted: 09/02/2012] [Indexed: 01/18/2023] Open
Abstract
We previously showed tumor-associated macrophages/microglia (TAMs) polarized to the M2 phenotype were significantly involved in tumor cell proliferation and poor clinical prognosis in patients with high grade gliomas. However, the detailed molecular mechanisms involved in the interaction between TAMs and tumor cells have been unclear. Current results reveal that, in coculture with human macrophages, BrdU incorporation was significantly elevated in glioma cells, and signal transducer and activator of transcription-3 (Stat3) activation was found in both cell types. Direct mixed coculture led to stronger Stat3 activation in tumor cells than did indirect separate coculture in Transwell chamber dishes. Screening with an array kit for phospho-receptor tyrosine kinases revealed that phosphorylation of macrophage-colony stimulating factor receptor (M-CSFR, CD115, or c-fms) is possibly involved in this cell-cell interaction; M-CSFR activation was detected in both cell types. Coculture-induced tumor cell activation was suppressed by siRNA-mediated downregulation of the M-CSFR in macrophages and by an inhibitor of M-CSFR (GW2580). Immunohistochemical analysis of phosphorylated (p)M-CSFR, pStat3, M-CSF, M2 ratio, and MIB-1(%) in high grade gliomas revealed that higher staining of pM-CSFR in tumor cells was significantly associated with higher M-CSF expression and higher MIB-1(%). Higher staining of pStat3 was associated with higher MIB-1(%). High M2 ratios were closely correlated with high MIB-1(%) and poor clinical prognosis. Targeting these molecules or deactivating M2 macrophages might be useful therapeutic strategies for high grade glioma patients.
Collapse
Affiliation(s)
- Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
450
|
Singh SK, Vartanian A, Burrell K, Zadeh G. A microRNA Link to Glioblastoma Heterogeneity. Cancers (Basel) 2012; 4:846-72. [PMID: 24213470 PMCID: PMC3712712 DOI: 10.3390/cancers4030846] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/28/2012] [Accepted: 08/21/2012] [Indexed: 12/18/2022] Open
Abstract
Glioblastomas (GBM) are one of the most malignant adult primary brain tumors. Through decades of research using various model systems and GBM patients, we have gained considerable insights into the mechanisms regulating GBM pathogenesis, but have mostly failed to significantly improve clinical outcome. For the most part GBM heterogeneity is responsible for this lack of progress. Here, we have discussed sources of cellular and microenvironmental heterogeneity in GBMs and their potential regulation through microRNA mediated mechanisms. We have focused on the role of individual microRNAs (miRNA) through their specific targets and miRNA mediated RNA-RNA interaction networks with the potential to influence various aspects of GBM heterogeneity including tumor neo-vascularization. We believe a better understanding of such mechanisms for regulation of GBM pathogenesis will be instrumental for future therapeutic options.
Collapse
Affiliation(s)
- Sanjay K Singh
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 1L7, Canada.
| | | | | | | |
Collapse
|