101
|
De I, Steffen MD, Clark PA, Patros CJ, Sokn E, Bishop SM, Litscher S, Maklakova VI, Kuo JS, Rodriguez FJ, Collier LS. CSF1 Overexpression Promotes High-Grade Glioma Formation without Impacting the Polarization Status of Glioma-Associated Microglia and Macrophages. Cancer Res 2016; 76:2552-60. [PMID: 27013192 DOI: 10.1158/0008-5472.can-15-2386] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/22/2016] [Indexed: 12/11/2022]
Abstract
Current therapies for high-grade gliomas extend survival only modestly. The glioma microenvironment, including glioma-associated microglia/macrophages (GAM), is a potential therapeutic target. The microglia/macrophage cytokine CSF1 and its receptor CSF1R are overexpressed in human high-grade gliomas. To determine whether the other known CSF1R ligand IL34 is expressed in gliomas, we examined expression array data of human high-grade gliomas and performed RT-PCR on glioblastoma sphere-forming cell lines (GSC). Expression microarray analyses indicated that CSF1, but not IL34, is frequently overexpressed in human tumors. We found that while GSCs did express CSF1, most GSC lines did not express detectable levels of IL34 mRNA. We therefore studied the impact of modulating CSF1 levels on gliomagenesis in the context of the GFAP-V12Ha-ras-IRESLacZ (Ras*) model. Csf1 deficiency deterred glioma formation in the Ras* model, whereas CSF1 transgenic overexpression decreased the survival of Ras* mice and promoted the formation of high-grade gliomas. Conversely, CSF1 overexpression increased GAM density, but did not impact GAM polarization state. Regardless of CSF1 expression status, most GAMs were negative for the M2 polarization markers ARG1 and CD206; when present, ARG1(+) and CD206(+) cells were found in regions of peripheral immune cell invasion. Therefore, our findings indicate that CSF1 signaling is oncogenic during gliomagenesis through a mechanism distinct from modulating GAM polarization status. Cancer Res; 76(9); 2552-60. ©2016 AACR.
Collapse
Affiliation(s)
- Ishani De
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Megan D Steffen
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul A Clark
- Department of Neurological Surgery and Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Clayton J Patros
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Emily Sokn
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Stephanie M Bishop
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Suzanne Litscher
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Vilena I Maklakova
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - John S Kuo
- Department of Neurological Surgery and Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Fausto J Rodriguez
- Division of Neuropathology, Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Lara S Collier
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
102
|
Domingues P, González-Tablas M, Otero Á, Pascual D, Miranda D, Ruiz L, Sousa P, Ciudad J, Gonçalves JM, Lopes MC, Orfao A, Tabernero MD. Tumor infiltrating immune cells in gliomas and meningiomas. Brain Behav Immun 2016. [PMID: 26216710 DOI: 10.1016/j.bbi.2015.07.019] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tumor-infiltrating immune cells are part of a complex microenvironment that promotes and/or regulates tumor development and growth. Depending on the type of cells and their functional interactions, immune cells may play a key role in suppressing the tumor or in providing support for tumor growth, with relevant effects on patient behavior. In recent years, important advances have been achieved in the characterization of immune cell infiltrates in central nervous system (CNS) tumors, but their role in tumorigenesis and patient behavior still remain poorly understood. Overall, these studies have shown significant but variable levels of infiltration of CNS tumors by macrophage/microglial cells (TAM) and to a less extent also lymphocytes (particularly T-cells and NK cells, and less frequently also B-cells). Of note, TAM infiltrate gliomas at moderate numbers where they frequently show an immune suppressive phenotype and functional behavior; in contrast, infiltration by TAM may be very pronounced in meningiomas, particularly in cases that carry isolated monosomy 22, where the immune infiltrates also contain greater numbers of cytotoxic T and NK-cells associated with an enhanced anti-tumoral immune response. In line with this, the presence of regulatory T cells, is usually limited to a small fraction of all meningiomas, while frequently found in gliomas. Despite these differences between gliomas and meningiomas, both tumors show heterogeneous levels of infiltration by immune cells with variable functionality. In this review we summarize current knowledge about tumor-infiltrating immune cells in the two most common types of CNS tumors-gliomas and meningiomas-, as well as the role that such immune cells may play in the tumor microenvironment in controlling and/or promoting tumor development, growth and control.
Collapse
Affiliation(s)
- Patrícia Domingues
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María González-Tablas
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Álvaro Otero
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Daniel Pascual
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - David Miranda
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Laura Ruiz
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Pablo Sousa
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Juana Ciudad
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | | | - María Celeste Lopes
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Alberto Orfao
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María Dolores Tabernero
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain; Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain; Instituto de Estudios de Ciencias de la salud de Castilla y León (IECSCYL-IBSAL) and Research Unit of the University Hospital of Salamanca, Salamanca, Spain.
| |
Collapse
|
103
|
Abstract
The dogma of the central nervous system (CNS) as an immune-privileged site has been substantially revised in recent years. CNS is an immunocompetent organ and actively interacts with the immune system. Microglia plays a leading role in a CNS immune response. However, in malignant gliomas, there is M2-polarization of microglia acquiring immunosuppressive and tumor-supportive properties. It occurs under the influence of tumor cytokines, such as transforming growth factor-β, interleukin-10, and prostaglandin E2. M2-polarized microglia exhibits reduced phagocytic activity, changes in the expression of many cellular determinants, or inverse of their functions, STAT3 activation, and production of immunosuppressive cytokines that suppress the function of cytotoxic CD8+ T cells or CD4+ T-helper cells type I. Myeloid-derived suppressor cells and regulatory T-lymphocytes, which have been recruited from peripheral blood into tumor tissue, also have immunosuppressive properties. The development of new treatment options for malignant gliomas must consider the role of the microenvironment in maintaining tumor vitality and progression.
Collapse
Affiliation(s)
- K E Borisov
- Republican Clinical Oncology Dispensary, Ministry of Health of the Republic of Bashkortostan, Ufa, Russia
| | - D D Sakaeva
- Republican Clinical Oncology Dispensary, Ministry of Health of the Republic of Bashkortostan, Ufa, Russia
| |
Collapse
|
104
|
Abstract
Gliomas are the most common primary brain tumors of the central nervous system, and carry a grim prognosis. Novel approaches utilizing the immune system as adjuvant therapy are quickly emerging as viable and effective options. Immunotherapeutic strategies being investigated to treat glioblastoma include: vaccination therapy targeted against either specific tumor antigens or whole tumor lysate, adoptive cellular therapy with cytotoxic T lymphocytes, chimeric antigen receptors and bi-specific T-cell engaging antibodies allowing circumvention of major histocompatibility complex restriction, aptamer therapy with aims for more efficient target delivery, and checkpoint blockade in order to release the tumor-mediated inhibition of the immune system. Given the heterogeneity of glioblastoma and its ability to gain mutations throughout the disease course, multifaceted treatment strategies utilizing multiple forms of immunotherapy in combination with conventional therapy will be most likely to succeed moving forward.
Collapse
Affiliation(s)
- Brandon D Liebelt
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Houston Methodist Neurological Institute, Houston, TX, USA
| | - Gaetano Finocchiaro
- Department of Neuro-oncology, IRCCS Istituto Neurologico Besta, Milan, Italy
| | - Amy B Heimberger
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
105
|
Wei J, Nduom EK, Kong LY, Hashimoto Y, Xu S, Gabrusiewicz K, Ling X, Huang N, Qiao W, Zhou S, Ivan C, Fuller GN, Gilbert MR, Overwijk W, Calin GA, Heimberger AB. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol 2015; 18:639-48. [PMID: 26658052 DOI: 10.1093/neuonc/nov292] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/31/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Antibody therapeutic targeting of the immune checkpoints cytotoxic T-lymphocyte-associated molecule 4 (CTLA-4) and programmed cell death 1 (PD-1) has demonstrated marked tumor regression in clinical trials. MicroRNAs (miRNAs) can modulate multiple gene transcripts including possibly more than one immune checkpoint and could be exploited as immune therapeutics. METHODS Using online miRNA targeting prediction algorithms, we searched for miRNAs that were predicted to target both PD-1 and CTLA-4. MiR-138 emerged as a leading candidate. The effects of miR-138 on CTLA-4 and PD-1 expression and function in T cells were determined and the therapeutic effect of intravenous administration of miR-138 was investigated in both immune-competent and -incompetent murine models of GL261 glioma. RESULTS Target binding algorithms predicted that miR-138 could bind the 3' untranslated regions of CTLA-4 and PD-1, which was confirmed with luciferase expression assays. Transfection of human CD4+ T cells with miR-138 suppressed expression of CTLA-4, PD-1, and Forkhead box protein 3 (FoxP3) in transfected human CD4+ T cells. In vivo miR-138 treatment of GL261 gliomas in immune-competent mice demonstrated marked tumor regression, a 43% increase in median survival time (P = .011), and an associated decrease in intratumoral FoxP3+ regulatory T cells, CTLA-4, and PD-1 expression. This treatment effect was lost in nude immune-incompetent mice and with depletion of CD4+ or CD8+ T cells, and miR-138 had no suppressive effect on glioma cells when treated directly at physiological in vivo doses. CONCLUSIONS MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints which may have rapid translational potential as a novel immunotherapeutic agent.
Collapse
Affiliation(s)
- Jun Wei
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Edjah K Nduom
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Yuuri Hashimoto
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Shuo Xu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Konrad Gabrusiewicz
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Xiaoyang Ling
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Neal Huang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Wei Qiao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Shouhao Zhou
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Cristina Ivan
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Greg N Fuller
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Mark R Gilbert
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Willem Overwijk
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - George A Calin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas (J.W., E.K.N., L.-Y.K., Y.H., S.X., K.G., X.L., N.H., A.B.H.); Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.Q., S.Z.); Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas (C.I.); Departments of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.N.F.); Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G.); Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas (W.O.); Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas (G.A.C.); Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China (S.X.)
| |
Collapse
|
106
|
Abstract
Glioblastomas (GBM) are one of the most recalcitrant brain tumors because of their aggressive invasive growth and resistance to therapy. They are highly heterogeneous malignancies at both the molecular and histological levels. Specific histological hallmarks including pseudopalisading necrosis and microvascular proliferation distinguish GBM from lower-grade gliomas, and make GBM one of the most hypoxic as well as angiogenic tumors. These microanatomical compartments present specific niches within the tumor microenvironment that regulate metabolic needs, immune surveillance, survival, invasion as well as cancer stem cell maintenance. Here we review features and functions of the distinct GBM niches, detail the different cell constituents and the functional status of the vasculature, and discuss prospects of therapeutically targeting GBM niche constituents.
Collapse
Affiliation(s)
- Dolores Hambardzumyan
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Gabriele Bergers
- Department of Neurological Surgery, University of California San Francisco, Helen Diller Family Cancer Research Center, 1450 3 Street, San Francisco, California 94158, USA; Brain Tumor Center, University of California San Francisco, Helen Diller Family Cancer Research Center, 1450 3 Street, San Francisco, California 94158, USA; UCSF Comprehensive Cancer Center, University of California San Francisco, Helen Diller Family Cancer Research Center, 1450 3 Street, San Francisco, California 94158, USA
| |
Collapse
|
107
|
Jha MK, Lee WH, Suk K. Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders. Biochem Pharmacol 2015; 103:1-16. [PMID: 26556658 DOI: 10.1016/j.bcp.2015.11.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
Abstract
Recent neuroscience research has established the adult brain as a dynamic organ having a unique ability to undergo changes with time. Neuroglia, especially microglia and astrocytes, provide dynamicity to the brain. Activation of these glial cells is a major component of the neuroinflammatory responses underlying brain injury and neurodegeneration. Glial cells execute functional reaction programs in response to diverse microenvironmental signals manifested by neuropathological conditions. Activated microglia exist along a continuum of two functional states of polarization namely M1-type (classical/proinflammatory activation) and M2-type (alternative/anti-inflammatory activation) as in macrophages. The balance between classically and alternatively activated microglial phenotypes influences disease progression in the CNS. The classically activated state of microglia drives the neuroinflammatory response and mediates the detrimental effects on neurons, whereas in their alternative activation state, which is apparently a beneficial activation state, the microglia play a crucial role in tissue maintenance and repair. Likewise, in response to immune or inflammatory microenvironments astrocytes also adopt neurotoxic or neuroprotective phenotypes. Reactive astrocytes exhibit two distinctive functional phenotypes defined by pro- or anti-inflammatory gene expression profile. In this review, we have thoroughly covered recent advances in the understanding of the functional polarization of brain and peripheral glia and its implications in neuroinflammation and neurological disorders. The identifiable phenotypes adopted by neuroglia in response to specific insult or injury can be exploited as promising diagnostic markers of neuroinflammatory diseases. Furthermore, harnessing the beneficial effects of the polarized glia could undoubtedly pave the way for the formulation of novel glia-based therapeutic strategies for diverse neurological disorders.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
108
|
Butowski N, Colman H, De Groot JF, Omuro AM, Nayak L, Wen PY, Cloughesy TF, Marimuthu A, Haidar S, Perry A, Huse J, Phillips J, West BL, Nolop KB, Hsu HH, Ligon KL, Molinaro AM, Prados M. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro Oncol 2015; 18:557-64. [PMID: 26449250 DOI: 10.1093/neuonc/nov245] [Citation(s) in RCA: 450] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/03/2015] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The colony stimulating factor 1 receptor (CSF1R) ligands, CSF1 and interleukin-34, and the KIT ligand, stem cell factor, are expressed in glioblastoma (GB). Microglia, macrophages, blood vessels, and tumor cells also express CSF1R, and depletion of the microglia reduces tumor burden and invasive capacity. PLX3397 is an oral, small molecule that selectively inhibits CSF1R and KIT, penetrates the blood-brain barrier in model systems, and represents a novel approach for clinical development. METHODS We conducted a phase II study in patients with recurrent GB. The primary endpoint was 6-month progression-free survival (PFS6). Secondary endpoints included overall survival response rate, safety, and plasma/tumor tissue pharmacokinetics. Exploratory endpoints included pharmacodynamic measures of drug effect in blood and tumor tissue. RESULTS A total of 37 patients were enrolled, with 13 treated prior to a planned surgical resection (Cohort 1) and 24 treated without surgery (Cohort 2). PLX3397 was given at an oral dose of 1000 mg daily and was well tolerated. The primary efficacy endpoint of PFS6 was only 8.6%, with no objective responses. Pharmacokinetic endpoints revealed a median maximal concentration (Cmax) of 8090 ng/mL, with a time to attain Cmax of 2 hour in plasma. Tumor tissue obtained after 7 days of drug exposure revealed a median drug level of 5500 ng/g. Pharmacodynamic changes included an increase in colony stimulating factor 1 and reduced CD14(dim)/CD16+ monocytes in plasma compared with pretreatment baseline values. CONCLUSION PLX3397 was well tolerated and readily crossed the blood-tumor barrier but showed no efficacy. Additional studies are ongoing, testing combination strategies and potential biomarkers to identify patients with greater likelihood of response.
Collapse
Affiliation(s)
- Nicholas Butowski
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Howard Colman
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - John F De Groot
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Antonio M Omuro
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Lakshmi Nayak
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Patrick Y Wen
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Timothy F Cloughesy
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Adhirai Marimuthu
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Sam Haidar
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Arie Perry
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Jason Huse
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Joanna Phillips
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Brian L West
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Keith B Nolop
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Henry H Hsu
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Keith L Ligon
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Annette M Molinaro
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| | - Michael Prados
- University of California at San Francisco, San Francisco, California (N.B., A.P., J.P., A.M.M, M.P.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah (H.C.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.F.D.G.); Department of Neurology, Memorial Sloan Kettering Cancer Hospital, New York, New York (A.M.O.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (L.N., P.Y.W., S.H., K.L.L.); UCLA Medical Center, Los Angeles, California (T.F.C.); Plexxikon Inc., Berkeley, California (A.M., B.L.W., K.B.N., H.H.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Pathology, Memorial Sloan Kettering Cancer Hospital, New York, New York (J.H.)
| |
Collapse
|
109
|
Differential gene methylation in paired glioblastomas suggests a role of immune response pathways in tumor progression. J Neurooncol 2015. [DOI: 10.1007/s11060-015-1869-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
110
|
Martín-Moreno AM, Roncador G, Maestre L, Mata E, Jiménez S, Martínez-Torrecuadrada JL, Reyes-García AI, Rubio C, Tomás JF, Estévez M, Pulford K, Piris MA, García JF. CSF1R Protein Expression in Reactive Lymphoid Tissues and Lymphoma: Its Relevance in Classical Hodgkin Lymphoma. PLoS One 2015; 10:e0125203. [PMID: 26066800 PMCID: PMC4466308 DOI: 10.1371/journal.pone.0125203] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 03/11/2015] [Indexed: 11/28/2022] Open
Abstract
Tumour-associated macrophages (TAMs) have been associated with survival in classic Hodgkin lymphoma (cHL) and other lymphoma types. The maturation and differentiation of tissue macrophages depends upon interactions between colony-stimulating factor 1 receptor (CSF1R) and its ligands. There remains, however, a lack of consistent information on CSF1R expression in TAMs. A new monoclonal antibody, FER216, was generated to investigate CSF1R protein distribution in formalin fixed tissue samples from 24 reactive lymphoid tissues and 187 different lymphoma types. We also analysed the distribution of CSF1R+, CD68+ and CD163+ macrophages by double immunostaining, and studied the relationship between CSF1R expression and survival in an independent series of 249 cHL patients. CSF1R+ TAMs were less frequent in B-cell lymphocytic leukaemia and lymphoblastic B-cell lymphoma than in diffuse large B-cell lymphoma, peripheral T-cell lymphoma, angioimmunoblastic T-cell lymphoma and cHL. HRS cells in cHL and, with the exception of three cases of anaplastic large cell lymphoma, the neoplastic cells in NHLs, lacked detectable CSF1R protein. A CSF1R+ enriched microenvironment in cHL was associated with shorter survival in an independent series of 249 cHL patients. CSF1R pathway activation was evident in the cHL and inactivation of this pathway could be a potential therapeutic target in cHL cases.
Collapse
Affiliation(s)
| | - Giovanna Roncador
- Monoclonal Antibodies Unit, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Lorena Maestre
- Monoclonal Antibodies Unit, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Elena Mata
- Pathology Department, MD Anderson Cancer Center, Madrid, Spain
| | - Scherezade Jiménez
- Monoclonal Antibodies Unit, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | | | - Ana I. Reyes-García
- Monoclonal Antibodies Unit, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Carmen Rubio
- Pathology Department, MD Anderson Cancer Center, Madrid, Spain
| | - José F. Tomás
- Haematology Department, MD Anderson Cancer Center, Madrid, Spain
| | - Mónica Estévez
- Haematology Department, MD Anderson Cancer Center, Madrid, Spain
| | - Karen Pulford
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Miguel A. Piris
- Pathology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Juan F. García
- Pathology Department, MD Anderson Cancer Center, Madrid, Spain
- * E-mail:
| |
Collapse
|
111
|
Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat Cell Biol 2015; 17:170-82. [PMID: 25580734 PMCID: PMC4312504 DOI: 10.1038/ncb3090] [Citation(s) in RCA: 691] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 11/26/2014] [Indexed: 02/06/2023]
Abstract
Tumor-associated macrophages (TAMs) are enriched in glioblastoma (GBM) that contains glioma stem cells (GSCs) at the apex of its cellular hierarchy. The correlation between TAM density and glioma grade suggests a supportive role of TAMs in tumor progression. Here we interrogated the molecular link between GSCs and TAM recruitment in GBMs and demonstrated that GSCs secrete Periostin (POSTN) to recruit TAMs. TAM density correlates with POSTN levels in human GBMs. Silencing POSTN in GSCs markedly reduced TAM density, inhibited tumor growth, and increased survival of mice bearing GSC-derived xenografts. We found that TAMs in GBMs are not brain-resident microglia, but mainly monocyte-derived macrophages from peripheral blood. Disrupting POSTN specifically attenuated the tumor supportive M2 type of TAMs in xenografts. POSTN recruits TAMs through integrin αvβ3 as blocking this signaling by an RGD peptide inhibited TAM recruitment. Our findings highlight the possibility of improving GBM treatment by targeting POSTN-mediated TAM recruitment.
Collapse
|
112
|
Huen SC, Huynh L, Marlier A, Lee Y, Moeckel GW, Cantley LG. GM-CSF Promotes Macrophage Alternative Activation after Renal Ischemia/Reperfusion Injury. J Am Soc Nephrol 2014; 26:1334-45. [PMID: 25388222 DOI: 10.1681/asn.2014060612] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/09/2014] [Indexed: 11/03/2022] Open
Abstract
After kidney ischemia/reperfusion (I/R) injury, monocytes home to the kidney and differentiate into activated macrophages. Whereas proinflammatory macrophages contribute to the initial kidney damage, an alternatively activated phenotype can promote normal renal repair. The microenvironment of the kidney during the repair phase mediates the transition of macrophage activation from a proinflammatory to a reparative phenotype. In this study, we show that macrophages isolated from murine kidneys during the tubular repair phase after I/R exhibit an alternative activation gene profile that differs from the canonical alternative activation induced by IL-4-stimulated STAT6 signaling. This unique activation profile can be reproduced in vitro by stimulation of bone marrow-derived macrophages with conditioned media from serum-starved mouse proximal tubule cells. Secreted tubular factors were found to activate macrophage STAT3 and STAT5 but not STAT6, leading to induction of the unique alternative activation pattern. Using STAT3-deficient bone marrow-derived macrophages and pharmacologic inhibition of STAT5, we found that tubular cell-mediated macrophage alternative activation is regulated by STAT5 activation. Both in vitro and after renal I/R, tubular cells expressed GM-CSF, a known STAT5 activator, and this pathway was required for in vitro alternative activation of macrophages by tubular cells. Furthermore, administration of a neutralizing antibody against GM-CSF after renal I/R attenuated kidney macrophage alternative activation and suppressed tubular proliferation. Taken together, these data show that tubular cells can instruct macrophage activation by secreting GM-CSF, leading to a unique macrophage reparative phenotype that supports tubular proliferation after sterile ischemic injury.
Collapse
Affiliation(s)
| | | | | | - Yashang Lee
- Section of Nephrology, Department of Medicine
| | - Gilbert W Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
113
|
Mechanisms of intimate and long-distance cross-talk between glioma and myeloid cells: how to break a vicious cycle. Biochim Biophys Acta Rev Cancer 2014; 1846:560-75. [PMID: 25453365 DOI: 10.1016/j.bbcan.2014.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/12/2014] [Accepted: 10/13/2014] [Indexed: 12/16/2022]
Abstract
Glioma-associated microglia and macrophages (GAMs) and myeloid-derived suppressor cells (MDSCs) condition the glioma microenvironment to generate an immunosuppressed niche for tumour expansion. This immunosuppressive microenvironment is considered to be shaped through a complex multi-step interactive process between glioma cells, GAMs and MDSCs. Glioma cells recruit GAMs and MDSCs to the tumour site and block their maturation. Glioma cell-derived factors subsequently skew these cells towards an immunosuppressive, tumour-promoting phenotype. Finally, GAMs and MDSCs enhance immune suppression in the glioma microenvironment and promote glioma growth, invasiveness, and neovascularization. The local and distant cross-talk between glioma cells and GAMs and MDSCs is regulated by a plethora of soluble proteins and cell surface-bound factors, and possibly via extracellular vesicles and platelets. Importantly, GAMs and MDSCs have been reported to impair the efficacy of glioma therapy, in particular immunotherapeutic approaches. Therefore, advancing our understanding of the function of GAMs and MDSCs in brain tumours and targeted intervention of their immunosuppressive function may benefit the treatment of glioma.
Collapse
|
114
|
Glass R, Synowitz M. CNS macrophages and peripheral myeloid cells in brain tumours. Acta Neuropathol 2014; 128:347-62. [PMID: 24722970 DOI: 10.1007/s00401-014-1274-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/10/2014] [Accepted: 03/25/2014] [Indexed: 12/15/2022]
Abstract
Primary brain tumours (gliomas) initiate a strong host response and can contain large amounts of immune cells (myeloid cells) such as microglia and tumour-infiltrating macrophages. In gliomas the course of pathology is not only controlled by the genetic make-up of the tumour cells, but also depends on the interplay with myeloid cells in the tumour microenvironment. Especially malignant gliomas such as glioblastoma multiforme (GBM) are notoriously immune-suppressive and it is now evident that GBM cells manipulate myeloid cells to support tumour expansion. The protumorigenic effects of glioma-associated myeloid cells comprise a support for angiogenesis as well as tumour cell invasion, proliferation and survival. Different strategies for inhibiting the pathological functions of myeloid cells in gliomas are explored, and blocking the tropism of microglia/macrophages to gliomas or manipulating the signal transduction pathways for immune cell activation has been successful in pre-clinical models. Hence, myeloid cells are now emerging as a promising target for new adjuvant therapies for gliomas. However, it is also becoming evident that some myeloid-directed glioma therapies may only be beneficial for distinct subclasses of gliomas and that a more cell-type-specific manipulation of either microglia or macrophages may improve therapeutic outcomes.
Collapse
|
115
|
Samaranayake HD, Pikkarainen JT, Wirth T, Stedt H, Lesch HP, Dragneva G, Vuorio T, Määttä AM, Airenne K, Ylä-Herttuala S. Soluble vascular endothelial growth factor receptor-1 improves therapeutic efficacy of suicide gene therapy in an angiogenesis-independent manner. Hum Gene Ther 2014; 25:942-54. [PMID: 25072110 DOI: 10.1089/hum.2013.191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Abstract Malignant gliomas (MGs) are highly vascularized, aggressive brain cancers carrying a dismal prognosis. Because of their high vascularity, anti-angiogenic therapy is a potential treatment option. Indeed, the anti-vascular endothelial growth factor (VEGF) antibody bevacizumab has demonstrated promising results in clinical trials. Similarly, adenovirus-medicated Herpes simplex virus thymidine kinase and ganciclovir (AdHSV-tk/GCV) suicide gene therapy has established itself in clinical trials as a potential novel therapeutic strategy for MGs. In this study, we demonstrate the feasibility of combining adenovirus-mediated soluble VEGF receptor-1 anti-angiogenic gene therapy with AdHSV-tk/GCV suicide gene therapy to treat experimental MGs. Our results reveal that, apart from inhibiting angiogenesis, other anti-tumor mechanisms, such as reduction of infiltration by tumor-associated macrophages/microglia, may contribute to the improved therapeutic benefit of combination therapy.
Collapse
Affiliation(s)
- Haritha D Samaranayake
- 1 Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland , FI-70211 Kuopio, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Bashir MR, Bhatti L, Marin D, Nelson RC. Emerging applications for ferumoxytol as a contrast agent in MRI. J Magn Reson Imaging 2014; 41:884-98. [PMID: 24974785 DOI: 10.1002/jmri.24691] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/18/2014] [Indexed: 12/12/2022] Open
Abstract
Ferumoxytol is an ultrasmall superparamagnetic iron oxide (USPIO) agent initially approved by the Food and Drug Administration (FDA) as an iron replacement therapy for patients with anemia due to chronic renal failure. Recently, ferumoxytol has been investigated extensively as an intravenous contrast agent in magnetic resonance imaging (MRI). Since it causes regional T1 and T2 * shortening in vivo, conventional pulse sequences can be used following ferumoxytol administration to demonstrate signal enhancement or loss. Ferumoxytol can be administered as a rapid bolus and has a long intravascular half-life on the order of 14-15 hours, making it a potentially useful agent for vascular and perfusion-weighted MRI. In comparison to other USPIOs, ferumoxytol is less limited by allergic and idiosyncratic reactions. Furthermore, since ferumoxytol is an iron-based agent with no potential for causing nephrogenic systemic fibrosis, it may be useful as an alternative to gadolinium-based contrast agents in patients with compromised renal function. Ferumoxytol is ultimately taken up by macrophages/the reticuloendothelial system in the liver, spleen, and lymph nodes, and this uptake mechanism is being explored as a novel imaging technique for vascular lesions, tumors, and lymph nodes. This article reviews the properties of ferumoxytol relevant to MRI as well as many of the uses for the agent currently under investigation.
Collapse
Affiliation(s)
- Mustafa R Bashir
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | |
Collapse
|
117
|
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: 27] [Impact Index Per Article: 2.5] [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
|
118
|
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: 12.2] [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
|