Tumor Microenvironment in the Brain
Abstract
:1. Introduction
2. Interactions between Blood Vessels and Cancer Cells in the Brain
2.1. Neovascularization in Brain Tumors
2.2. Neovascularization-Independent Contribution of Endothelial Cells to the Brain Tumor Progression
3. Pericytes
The Role of MSCs and Pericytes in Brain Tumor Growth
4. Contribution of Myeloid Cells to Brain Tumors
4.1. Macrophages and Microglia in the Central Nervous System
4.2. Microglia and Macrophages in Primary and Metastatic Brain Tumors
4.3. Functional Role of Myeloid Cells in the Progression of Brain Tumors in Animal Models
4.4. Microglia/Macrophages Contribute to the Immunosuppressed Environment in the CNS
4.5. Myeloid Derived Suppressor Cells
5. Astrocytes
The Role of Astrocytes in Brain Tumors
6. Tumor Microenvironment in the Brain: Therapeutic Opportunities and Challenges
6.1. Targeting Angiogenesis
6.2. Cellular Vehicles for the Delivery of Therapeutic Agents to Brain Tumors
6.3. Other Potential Targets in Brain Tumor Microenvironment
7. Conclusions
Acknowledgements
Conflict of Interest
References
- Al-Shamy, G.; Sawaya, R. Management of brain metastases: The indispensable role of surgery. J. Neurooncol. 2009, 92, 275–282. [Google Scholar] [CrossRef]
- DeAngelis, L.M. Treatment of brain metastasis. J. Support. Oncol. 2008, 6, 87–88. [Google Scholar]
- Gavrilovic, I.T.; Posner, J.B. Brain metastases: Epidemiology and pathophysiology. J. Neurooncol. 2005, 75, 5–14. [Google Scholar] [CrossRef]
- Gerrard, G.E.; Franks, K.N. Overview of the diagnosis and management of brain, spine, and meningeal metastases. J. Neurol. Neurosurg. Psychiatr. 2004, 75, ii37–ii42. [Google Scholar] [CrossRef]
- Nikiforova, M.N.; Hamilton, R.L. Molecular diagnostics of gliomas. Arch. Pathol. Lab. Med. 2011, 135, 558–568. [Google Scholar]
- Davis, F.G.; Freels, S.; Grutsch, J.; Barlas, S.; Brem, S. Survival rates in patients with primary malignant brain tumors stratified by patient age and tumor histological type: An analysis based on Surveillance, Epidemiology, and End Results (SEER) data, 1973-1991. J. Neurosurg. 1998, 88, 1–10. [Google Scholar] [CrossRef]
- Davis, F.G.; McCarthy, B.J.; Berger, M.S. Centralized databases available for describing primary brain tumor incidence, survival, and treatment: Central Brain Tumor Registry of the United States; Surveillance, Epidemiology, and End Results; and National Cancer Data Base. Neuro-Oncology 1999, 1, 205–211. [Google Scholar]
- Ballman, K.V.; Buckner, J.C.; Brown, P.D.; Giannini, C.; Flynn, P.J.; LaPlant, B.R.; Jaeckle, K.A. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro-Oncology 2007, 9, 29–38. [Google Scholar]
- Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-Year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef]
- Wong, E.T.; Hess, K.R.; Gleason, M.J.; Jaeckle, K.A.; Kyritsis, A.P.; Prados, M.D.; Levin, V.A.; Yung, W.K. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J. Clin. Oncol. 1999, 17, 2572–2578. [Google Scholar]
- Albini, A.; Sporn, M.B. The tumour microenvironment as a target for chemoprevention. Nat. Rev. Cancer 2007, 7, 139–147. [Google Scholar]
- Joyce, J.A.; Pollard, J.W. Microenvironmental regulation of metastasis. Nat. Rev. Cancer 2009, 9, 239–252. [Google Scholar] [CrossRef]
- Witkiewicz, A.K.; Casimiro, M.C.; Dasgupta, A.; Mercier, I.; Wang, C.; Bonuccelli, G.; Jasmin, J.F.; Frank, P.G.; Pestell, R.G.; Kleer, C.G.; et al. Towards a new “stromal-based” classification system for human breast cancer prognosis and therapy. Cell Cycle 2009, 8, 1654–1658. [Google Scholar]
- Nicolson, G.L.; Menter, D.G.; Herrmann, J.L.; Yun, Z.; Cavanaugh, P.; Marchetti, D. Brain metastasis: Role of trophic, autocrine, and paracrine factors in tumor invasion and colonization of the central nervous system. Curr. Top. Microbiol. Immunol. 1996, 213, 89–115. [Google Scholar]
- Zhang, C.; Zhang, F.; Tsan, R.; Fidler, I.J. Transforming growth factor-beta2 is a molecular determinant for site-specific melanoma metastasis in the brain. Cancer Res. 2009, 69, 828–835. [Google Scholar]
- Blouw, B.; Song, H.; Tihan, T.; Bosze, J.; Ferrara, N.; Gerber, H.P.; Johnson, R.S.; Bergers, G. The hypoxic response of tumors is dependent on their microenvironment. Cancer Cell 2003, 4, 133–146. [Google Scholar] [CrossRef]
- Lorger, M.; Krueger, J.S.; O'Neal, M.; Staflin, K.; Felding-Habermann, B. Activation of tumor cell integrin alphavbeta3 controls angiogenesis and metastatic growth in the brain. Proc. Natl. Acad. Sci. USA 2009, 106, 10666–10671. [Google Scholar]
- Guo, P.; Xu, L.; Pan, S.; Brekken, R.A.; Yang, S.T.; Whitaker, G.B.; Nagane, M.; Thorpe, P.E.; Rosenbaum, J.S.; Su Huang, H.J.; et al. Vascular endothelial growth factor isoforms display distinct activities in promoting tumor angiogenesis at different anatomic sites. Cancer Res. 2001, 61, 8569–8577. [Google Scholar]
- Deli, M.A.; Abraham, C.S.; Kataoka, Y.; Niwa, M. Permeability studies on in vitro blood-brain barrier models: Physiology, pathology, and pharmacology. Cell. Mol. Neurobiol. 2005, 25, 59–127. [Google Scholar] [CrossRef]
- Lee, S.W.; Kim, W.J.; Park, J.A.; Choi, Y.K.; Kwon, Y.W.; Kim, K.W. Blood-brain barrier interfaces and brain tumors. Arch. Pharm. Res. 2006, 29, 265–275. [Google Scholar] [CrossRef]
- Nag, S. Morphology and molecular properties of cellular components of normal cerebral vessels. Methods Mol. Med. 2003, 89, 3–36. [Google Scholar]
- Carbonell, W.S.; Ansorge, O.; Sibson, N.; Muschel, R. The vascular basement membrane as “soil” in brain metastasis. PLoS One 2009, 4, e5857. [Google Scholar]
- Lorger, M.; Felding-Habermann, B. Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis. Am. J. Pathol. 2010, 176, 2958–2971. [Google Scholar] [CrossRef]
- Kienast, Y.; von Baumgarten, L.; Fuhrmann, M.; Klinkert, W.E.; Goldbrunner, R.; Herms, J.; Winkler, F. Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 2010, 16, 116–122. [Google Scholar]
- Fidler, I.J.; Yano, S.; Zhang, R.D.; Fujimaki, T.; Bucana, C.D. The seed and soil hypothesis: Vascularisation and brain metastases. Lancet Oncol. 2002, 3, 53–57. [Google Scholar] [CrossRef]
- Nir, I.; Levanon, D.; Iosilevsky, G. Permeability of blood vessels in experimental gliomas: Uptake of 99mTc-glucoheptonate and alteration in blood-brain barrier as determined by cytochemistry and electron microscopy. Neurosurgery 1989, 25, 523–532. [Google Scholar] [CrossRef]
- Zhang, R.D.; Price, J.E.; Fujimaki, T.; Bucana, C.D.; Fidler, I.J. Differential permeability of the blood-brain barrier in experimental brain metastases produced by human neoplasms implanted into nude mice. Am. J. Pathol. 1992, 141, 1115–1124. [Google Scholar]
- Lockman, P.R.; Mittapalli, R.K.; Taskar, K.S.; Rudraraju, V.; Gril, B.; Bohn, K.A.; Adkins, C.E.; Roberts, A.; Thorsheim, H.R.; Gaasch, J.A.; et al. Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clin. Cancer Res. 2010, 16, 5664–5678. [Google Scholar]
- Wen, P.Y.; Kesari, S. Malignant gliomas in adults. N. Engl. J. Med. 2008, 359, 492–507. [Google Scholar] [CrossRef]
- Chao, H.; Hirschi, K.K. Hemato-vascular origins of endothelial progenitor cells? Microvasc. Res. 2010, 79, 169–173. [Google Scholar] [CrossRef]
- Dome, B.; Dobos, J.; Tovari, J.; Paku, S.; Kovacs, G.; Ostoros, G.; Timar, J. Circulating bone marrow-derived endothelial progenitor cells: Characterization, mobilization, and therapeutic considerations in malignant disease. Cytometry A 2008, 73, 186–193. [Google Scholar]
- Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 2010, 468, 824–828. [Google Scholar]
- Bernstein, J.J.; Woodard, C.A. Glioblastoma cells do not intravasate into blood vessels. Neurosurgery 1995, 36, 124–132. [Google Scholar] [CrossRef]
- Holash, J.; Maisonpierre, P.C.; Compton, D.; Boland, P.; Alexander, C.R.; Zagzag, D.; Yancopoulos, G.D.; Wiegand, S.J. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 1999, 284, 1994–1998. [Google Scholar]
- Baeriswyl, V.; Christofori, G. The angiogenic switch in carcinogenesis. Semin. Cancer Biol. 2009, 19, 329–337. [Google Scholar] [CrossRef]
- Hanahan, D.; Folkman, J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996, 86, 353–364. [Google Scholar] [CrossRef]
- Harper, J.; Moses, M.A. Molecular regulation of tumor angiogenesis: Mechanisms and therapeutic implications. EXS 2006, 96, 223–268. [Google Scholar]
- Iruela-Arispe, M.L.; Dvorak, H.F. Angiogenesis: A dynamic balance of stimulators and inhibitors. Thromb. Haemost. 1997, 78, 672–677. [Google Scholar]
- Shweiki, D.; Neeman, M.; Itin, A.; Keshet, E. Induction of vascular endothelial growth factor expression by hypoxia and by glucose deficiency in multicell spheroids: Implications for tumor angiogenesis. Proc. Natl. Acad. Sci. USA 1995, 92, 768–772. [Google Scholar]
- Du, R.; Petritsch, C.; Lu, K.; Liu, P.; Haller, A.; Ganss, R.; Song, H.; Vandenberg, S.; Bergers, G. Matrix metalloproteinase-2 regulates vascular patterning and growth affecting tumor cell survival and invasion in GBM. Neuro-Oncology 2008, 10, 254–264. [Google Scholar] [CrossRef]
- Leenders, W.P.; Kusters, B.; Verrijp, K.; Maass, C.; Wesseling, P.; Heerschap, A.; Ruiter, D.; Ryan, A.; de Waal, R. Antiangiogenic therapy of cerebral melanoma metastases results in sustained tumor progression via vessel co-option. Clin. Cancer Res. 2004, 10, 6222–6230. [Google Scholar]
- Paez-Ribes, M.; Allen, E.; Hudock, J.; Takeda, T.; Okuyama, H.; Vinals, F.; Inoue, M.; Bergers, G.; Hanahan, D.; Casanovas, O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009, 15, 220–231. [Google Scholar] [CrossRef]
- Aicher, A.; Zeiher, A.M.; Dimmeler, S. Mobilizing endothelial progenitor cells. Hypertension 2005, 45, 321–325. [Google Scholar] [CrossRef]
- Bertolini, F.; Shaked, Y.; Mancuso, P.; Kerbel, R.S. The multifaceted circulating endothelial cell in cancer: Towards marker and target identification. Nat. Rev. Cancer 2006, 6, 835–845. [Google Scholar] [CrossRef]
- Rafat, N.; Beck, G.; Schulte, J.; Tuettenberg, J.; Vajkoczy, P. Circulating endothelial progenitor cells in malignant gliomas. J. Neurosurg. 2009, 112, 43–49. [Google Scholar]
- Zheng, P.P.; Hop, W.C.; Luider, T.M.; Sillevis Smitt, P.A.; Kros, J.M. Increased levels of circulating endothelial progenitor cells and circulating endothelial nitric oxide synthase in patients with gliomas. Ann. Neurol. 2007, 62, 40–48. [Google Scholar] [CrossRef]
- Aghi, M.; Cohen, K.S.; Klein, R.J.; Scadden, D.T.; Chiocca, E.A. Tumor stromal-derived factor-1 recruits vascular progenitors to mitotic neovasculature, where microenvironment influences their differentiated phenotypes. Cancer Res. 2006, 66, 9054–9064. [Google Scholar]
- de Palma, M.; Venneri, M.A.; Galli, R.; Sergi Sergi, L.; Politi, L.S.; Sampaolesi, M.; Naldini, L. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 2005, 8, 211–226. [Google Scholar] [CrossRef]
- Duda, D.G.; Cohen, K.S.; Kozin, S.V.; Perentes, J.Y.; Fukumura, D.; Scadden, D.T.; Jain, R.K. Evidence for incorporation of bone marrow-derived endothelial cells into perfused blood vessels in tumors. Blood 2006, 107, 2774–2776. [Google Scholar] [CrossRef]
- Machein, M.R.; Renninger, S.; de Lima-Hahn, E.; Plate, K.H. Minor contribution of bone marrow-derived endothelial progenitors to the vascularization of murine gliomas. Brain Pathol. 2003, 13, 582–597. [Google Scholar]
- Moore, X.L.; Lu, J.; Sun, L.; Zhu, C.J.; Tan, P.; Wong, M.C. Endothelial progenitor cells’ “homing” specificity to brain tumors. Gene Ther. 2004, 11, 811–818. [Google Scholar] [CrossRef]
- Santarelli, J.G.; Udani, V.; Yung, Y.C.; Cheshier, S.; Wagers, A.; Brekken, R.A.; Weissman, I.; Tse, V. Incorporation of bone marrow-derived Flk-1-expressing CD34+ cells in the endothelium of tumor vessels in the mouse brain. Neurosurgery 2006, 59, 374–382. [Google Scholar] [CrossRef]
- Udani, V.; Santarelli, J.; Yung, Y.; Cheshier, S.; Andrews, A.; Kasad, Z.; Tse, V. Differential expression of angiopoietin-1 and angiopoietin-2 may enhance recruitment of bone-marrow-derived endothelial precursor cells into brain tumors. Neurol. Res. 2005, 27, 801–806. [Google Scholar] [CrossRef]
- Bertolini, F.; Paul, S.; Mancuso, P.; Monestiroli, S.; Gobbi, A.; Shaked, Y.; Kerbel, R.S. Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells. Cancer Res. 2003, 63, 4342–4346. [Google Scholar]
- Shaked, Y.; Ciarrocchi, A.; Franco, M.; Lee, C.R.; Man, S.; Cheung, A.M.; Hicklin, D.J.; Chaplin, D.; Foster, F.S.; Benezra, R.; et al. Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Science 2006, 313, 1785–1787. [Google Scholar]
- Dome, B.; Timar, J.; Paku, S. A novel concept of glomeruloid body formation in experimental cerebral metastases. J. Neuropathol. Exp. Neurol. 2003, 62, 655–661. [Google Scholar]
- Winkler, F.; Kienast, Y.; Fuhrmann, M.; von Baumgarten, L.; Burgold, S.; Mitteregger, G.; Kretzschmar, H.; Herms, J. Imaging glioma cell invasion in vivo reveals mechanisms of dissemination and peritumoral angiogenesis. Glia 2009, 57, 1306–1315. [Google Scholar] [CrossRef]
- Zadeh, G.; Reti, R.; Koushan, K.; Baoping, Q.; Shannon, P.; Guha, A. Regulation of the pathological vasculature of malignant astrocytomas by angiopoietin-1. Neoplasia 2005, 7, 1081–1090. [Google Scholar] [CrossRef]
- Farin, A.; Suzuki, S.O.; Weiker, M.; Goldman, J.E.; Bruce, J.N.; Canoll, P. Transplanted glioma cells migrate and proliferate on host brain vasculature: A dynamic analysis. Glia 2006, 53, 799–808. [Google Scholar] [CrossRef]
- Calabrese, C.; Poppleton, H.; Kocak, M.; Hogg, T.L.; Fuller, C.; Hamner, B.; Oh, E.Y.; Gaber, M.W.; Finklestein, D.; Allen, M.; et al. A perivascular niche for brain tumor stem cells. Cancer Cell 2007, 11, 69–82. [Google Scholar] [CrossRef]
- Veeravagu, A.; Bababeygy, S.R.; Kalani, M.Y.; Hou, L.C.; Tse, V. The cancer stem cell-vascular niche complex in brain tumor formation. Stem Cells Dev. 2008, 17, 859–867. [Google Scholar] [CrossRef]
- Charles, N.; Ozawa, T.; Squatrito, M.; Bleau, A.M.; Brennan, C.W.; Hambardzumyan, D.; Holland, E.C. Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell 2010, 6, 141–152. [Google Scholar] [CrossRef]
- Bao, S.; Wu, Q.; Sathornsumetee, S.; Hao, Y.; Li, Z.; Hjelmeland, A.B.; Shi, Q.; McLendon, R.E.; Bigner, D.D.; Rich, J.N. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006, 66, 7843–7848. [Google Scholar]
- Bergers, G.; Song, S. The role of pericytes in blood-vessel formation and maintenance. Neuro-Oncology 2005, 7, 452–464. [Google Scholar] [CrossRef]
- Hellstrom, M.; Kalen, M.; Lindahl, P.; Abramsson, A.; Betsholtz, C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 1999, 126, 3047–3055. [Google Scholar]
- Leveen, P.; Pekny, M.; Gebre-Medhin, S.; Swolin, B.; Larsson, E.; Betsholtz, C. Mice deficient for PDGF B show renal, cardiovascular, and hematological abnormalities. Genes Dev. 1994, 8, 1875–1887. [Google Scholar] [CrossRef]
- Soriano, P. Abnormal kidney development and hematological disorders in PDGF beta-receptor mutant mice. Genes Dev. 1994, 8, 1888–1896. [Google Scholar] [CrossRef]
- Song, S.; Ewald, A.J.; Stallcup, W.; Werb, Z.; Bergers, G. PDGFRbeta+ perivascular progenitor cellsin tumours regulate pericyte differentiation and vascular survival. Nat. Cell Biol. 2005, 7, 870–879. [Google Scholar] [CrossRef]
- Huang, F.J.; You, W.K.; Bonaldo, P.; Seyfried, T.N.; Pasquale, E.B.; Stallcup, W.B. Pericyte deficiencies lead to aberrant tumor vascularizaton in the brain of the NG2 null mouse. Dev. Biol. 2010, 344, 1035–1046. [Google Scholar]
- Ozerdem, U.; Stallcup, W.B. Pathological angiogenesis is reduced by targeting pericytes via the NG2 proteoglycan. Angiogenesis 2004, 7, 269–276. [Google Scholar] [CrossRef]
- Fukushi, J.; Makagiansar, I.T.; Stallcup, W.B. NG2 proteoglycan promotes endothelial cell motility and angiogenesis via engagement of galectin-3 and alpha3beta1 integrin. Mol. Biol. Cell 2004, 15, 3580–3590. [Google Scholar] [CrossRef]
- Lamagna, C.; Bergers, G. The bone marrow constitutes a reservoir of pericyte progenitors. J. Leukoc. Biol. 2006, 80, 677–681. [Google Scholar] [CrossRef]
- Bababeygy, S.R.; Cheshier, S.H.; Hou, L.C.; Higgins, D.M.; Weissman, I.L.; Tse, V.C. Hematopoietic stem cell-derived pericytic cells in brain tumor angio-architecture. Stem Cells Dev. 2008, 17, 11–18. [Google Scholar] [CrossRef]
- Bexell, D.; Gunnarsson, S.; Tormin, A.; Darabi, A.; Gisselsson, D.; Roybon, L.; Scheding, S.; Bengzon, J. Bone marrow multipotent mesenchymal stroma cells act as pericyte-like migratory vehicles in experimental gliomas. Mol. Ther. 2009, 17, 183–190. [Google Scholar] [CrossRef]
- Winkler, E.A.; Bell, R.D.; Zlokovic, B.V. Central nervous system pericytes in health and disease. Nat. Neurosci. 2011, 14, 1398–1405. [Google Scholar]
- Armulik, A.; Genove, G.; Mae, M.; Nisancioglu, M.H.; Wallgard, E.; Niaudet, C.; He, L.; Norlin, J.; Lindblom, P.; Strittmatter, K.; et al. Pericytes regulate the blood-brain barrier. Nature 2010, 468, 557–561. [Google Scholar]
- Daneman, R.; Zhou, L.; Kebede, A.A.; Barres, B.A. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010, 468, 562–566. [Google Scholar]
- Birnbaum, T.; Hildebrandt, J.; Nuebling, G.; Sostak, P.; Straube, A. Glioblastoma-dependent differentiation and angiogenic potential of human mesenchymal stem cells in vitro. Neuro-Oncology 2011, 105, 57–65. [Google Scholar] [CrossRef]
- Nakamura, K.; Ito, Y.; Kawano, Y.; Kurozumi, K.; Kobune, M.; Tsuda, H.; Bizen, A.; Honmou, O.; Niitsu, Y.; Hamada, H. Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther. 2004, 11, 1155–1164. [Google Scholar] [CrossRef]
- Nakamizo, A.; Marini, F.; Amano, T.; Khan, A.; Studeny, M.; Gumin, J.; Chen, J.; Hentschel, S.; Vecil, G.; Dembinski, J.; et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005, 65, 3307–3318. [Google Scholar]
- Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegue, E.; Song, H.; Vandenberg, S.; Johnson, R.S.; Werb, Z.; et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 2008, 13, 206–220. [Google Scholar] [CrossRef]
- You, W.K.; Bonaldo, P.; Stallcup, W.B. Collagen VI ablation retards brain tumor progression due to deficits in assembly of the vascular basal lamina. Am. J. Pathol. 2012, 180, 1145–1158. [Google Scholar] [CrossRef]
- Geissmann, F.; Jung, S.; Littman, D.R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003, 19, 71–82. [Google Scholar] [CrossRef]
- Gordon, S.; Taylor, P.R. Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 2005, 5, 953–964. [Google Scholar] [CrossRef]
- Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef]
- Pollard, J.W. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer 2004, 4, 71–78. [Google Scholar] [CrossRef]
- Mantovani, A.; Sica, A. Macrophages, innate immunity and cancer: Balance, tolerance, and diversity. Curr. Opin. Immunol. 2010, 22, 231–237. [Google Scholar]
- Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar]
- Davis, E.J.; Foster, T.D.; Thomas, W.E. Cellular forms and functions of brain microglia. Brain Res. Bull. 1994, 34, 73–78. [Google Scholar] [CrossRef]
- Guillemin, G.J.; Brew, B.J. Microglia, macrophages, perivascular macrophages, and pericytes: A review of function and identification. J. Leukoc. Biol. 2004, 75, 388–397. [Google Scholar] [CrossRef]
- Hickey, W.F.; Kimura, H. Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo. Science 1988, 239, 290–292. [Google Scholar]
- Streit, W.J.; Conde, J.R.; Fendrick, S.E.; Flanary, B.E.; Mariani, C.L. Role of microglia in the central nervous system's immune response. Neurol. Res. 2005, 27, 685–691. [Google Scholar]
- Cartier, N.; Hacein-Bey-Abina, S.; Bartholomae, C.C.; Veres, G.; Schmidt, M.; Kutschera, I.; Vidaud, M.; Abel, U.; Dal-Cortivo, L.; Caccavelli, L.; et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 2009, 326, 818–823. [Google Scholar]
- Hess, D.C.; Abe, T.; Hill, W.D.; Studdard, A.M.; Carothers, J.; Masuya, M.; Fleming, P.A.; Drake, C.J.; Ogawa, M. Hematopoietic origin of microglial and perivascular cells in brain. Exp. Neurol. 2004, 186, 134–144. [Google Scholar] [CrossRef]
- Lesniak, M.S.; Kelleher, E.; Pardoll, D.; Cui, Y. Targeted gene therapy to antigen-presenting cells in the central nervous system using hematopoietic stem cells. Neurol. Res. 2005, 27, 820–826. [Google Scholar] [CrossRef]
- Priller, J.; Flugel, A.; Wehner, T.; Boentert, M.; Haas, C.A.; Prinz, M.; Fernandez-Klett, F.; Prass, K.; Bechmann, I.; de Boer, B.A.; et al. Targeting gene-modified hematopoietic cells to the central nervous system: Use of green fluorescent protein uncovers microglial engraftment. Nat. Med. 2001, 7, 1356–1361. [Google Scholar]
- Soulas, C.; Donahue, R.E.; Dunbar, C.E.; Persons, D.A.; Alvarez, X.; Williams, K.C. Genetically modified CD34+ hematopoietic stem cells contribute to turnover of brain perivascular macrophages in long-term repopulated primates. Am. J. Pathol. 2009, 174, 1808–1817. [Google Scholar] [CrossRef]
- Vallieres, L.; Sawchenko, P.E. Bone marrow-derived cells that populate the adult mouse brain preserve their hematopoietic identity. J. Neurosci. 2003, 23, 5197–5207. [Google Scholar]
- Biffi, A.; de Palma, M.; Quattrini, A.; Del Carro, U.; Amadio, S.; Visigalli, I.; Sessa, M.; Fasano, S.; Brambilla, R.; Marchesini, S.; et al. Correction of metachromatic leukodystrophy in the mouse model by transplantation of genetically modified hematopoietic stem cells. J. Clin. Invest. 2004, 113, 1118–1129. [Google Scholar]
- Getts, D.R.; Terry, R.L.; Getts, M.T.; Muller, M.; Rana, S.; Shrestha, B.; Radford, J.; van Rooijen, N.; Campbell, I.L.; King, N.J. Ly6c+ “inflammatory monocytes” are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. J. Exp. Med. 2008, 205, 2319–2337. [Google Scholar] [CrossRef]
- Yang, I.; Han, S.J.; Kaur, G.; Crane, C.; Parsa, A.T. The role of microglia in central nervous system immunity and glioma immunology. J. Clin. Neurosci. 2009, 17, 6–10. [Google Scholar]
- Kettenmann, H.; Hanisch, U.K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
- Davoust, N.; Vuaillat, C.; Androdias, G.; Nataf, S. From bone marrow to microglia: Barriers and avenues. Trends Immunol. 2008, 29, 227–234. [Google Scholar] [CrossRef]
- Sedgwick, J.D.; Schwender, S.; Imrich, H.; Dorries, R.; Butcher, G.W.; ter Meulen, V. Isolation and direct characterization of resident microglial cells from the normal and inflamed central nervous system. Proc. Natl. Acad. Sci. USA 1991, 88, 7438–7442. [Google Scholar]
- Bertolotto, A.; Agresti, C.; Castello, A.; Manzardo, E.; Riccio, A. 5D4 keratan sulfate epitope identifies a subset of ramified microglia in normal central nervous system parenchyma. J. Neuroimmunol. 1998, 85, 69–77. [Google Scholar] [CrossRef]
- Wilms, H.; Wollmer, M.A.; Sievers, J. In vitro-staining specificity of the antibody 5-D-4 for microglia but not for monocytes and macrophages indicates that microglia are a unique subgroup of the myelomonocytic lineage. J. Neuroimmunol. 1999, 98, 89–95. [Google Scholar] [CrossRef]
- Daginakatte, G.C.; Gutmann, D.H. Neurofibromatosis-1 (Nf1) heterozygous brain microglia elaborate paracrine factors that promote Nf1-deficient astrocyte and glioma growth. Hum. Mol. Genet. 2007, 16, 1098–1112. [Google Scholar] [CrossRef]
- Fitzgerald, D.P.; Palmieri, D.; Hua, E.; Hargrave, E.; Herring, J.M.; Qian, Y.; Vega-Valle, E.; Weil, R.J.; Stark, A.M.; Vortmeyer, A.O.; et al. Reactive glia are recruited by highly proliferative brain metastases of breast cancer and promote tumor cell colonization. Clin. Exp. Metastasis. 2008, 25, 799–810. [Google Scholar] [CrossRef]
- He, B.P.; Wang, J.J.; Zhang, X.; Wu, Y.; Wang, M.; Bay, B.H.; Chang, A.Y. Differential reactions of microglia to brain metastasis of lung cancer. Mol. Med. 2006, 12, 161–170. [Google Scholar] [CrossRef]
- Hoelzinger, D.B.; Demuth, T.; Berens, M.E. Autocrine factors that sustain glioma invasion and paracrine biology in the brain microenvironment. J. Natl. Cancer Inst. 2007, 99, 1583–1593. [Google Scholar] [CrossRef]
- Roggendorf, W.; Strupp, S.; Paulus, W. Distribution and characterization of microglia/macrophages in human brain tumors. Acta Neuropathol. 1996, 92, 288–293. [Google Scholar] [CrossRef]
- Zhang, M.; Olsson, Y. Reactions of astrocytes and microglial cells around hematogenous metastases of the human brain. Expression of endothelin-like immunoreactivity in reactive astrocytes and activation of microglial cells. J. Neurol. Sci. 1995, 134, 26–32. [Google Scholar] [CrossRef]
- Morantz, R.A.; Wood, G.W.; Foster, M.; Clark, M.; Gollahon, K. Macrophages in experimental and human brain tumors. Part 2: Studies of the macrophage content of human brain tumors. J. Neurosurg. 1979, 50, 305–311. [Google Scholar] [CrossRef]
- Morantz, R.A.; Wood, G.W.; Foster, M.; Clark, M.; Gollahon, K. Macrophages in experimental and human brain tumors. Part 1: Studies of the macrophage content of experimental rat brain tumors of varying immunogenicity. J. Neurosurg. 1979, 50, 298–304. [Google Scholar] [CrossRef]
- Badie, B.; Schartner, J.; Prabakaran, S.; Paul, J.; Vorpahl, J. Expression of Fas ligand by microglia: Possible role in glioma immune evasion. J. Neuroimmunol. 2001, 120, 19–24. [Google Scholar] [CrossRef]
- Klein, R.; Roggendorf, W. Increased microglia proliferation separates pilocytic astrocytomas from diffuse astrocytomas: A double labeling study. Acta Neuropathol. 2001, 101, 245–248. [Google Scholar]
- Badie, B.; Schartner, J. Role of microglia in glioma biology. Microsc. Res. Tech. 2001, 54, 106–113. [Google Scholar] [CrossRef]
- Kielian, T.; van Rooijen, N.; Hickey, W.F. MCP-1 expression in CNS-1 astrocytoma cells: Implications for macrophage infiltration into tumors in vivo. J. Neurooncol. 2002, 56, 1–12. [Google Scholar] [CrossRef]
- Leung, S.Y.; Wong, M.P.; Chung, L.P.; Chan, A.S.; Yuen, S.T. Monocyte chemoattractant protein-1 expression and macrophage infiltration in gliomas. Acta Neuropathol. 1997, 93, 518–527. [Google Scholar] [CrossRef]
- Mantovani, A.; Bottazzi, B.; Colotta, F.; Sozzani, S.; Ruco, L. The origin and function of tumor-associated macrophages. Immunol. Today 1992, 13, 265–270. [Google Scholar] [CrossRef]
- Okada, M.; Saio, M.; Kito, Y.; Ohe, N.; Yano, H.; Yoshimura, S.; Iwama, T.; Takami, T. Tumor-associated macrophage/microglia infiltration in human gliomas is correlated with MCP-3, but not MCP-1. Int. J. Oncol. 2009, 34, 1621–1627. [Google Scholar]
- Badie, B.; Schartner, J.; Klaver, J.; Vorpahl, J. In vitro modulation of microglia motility by glioma cells is mediated by hepatocyte growth factor/scatter factor. Neurosurgery 1999, 44, 1077–1082. [Google Scholar] [CrossRef]
- Nishie, A.; Ono, M.; Shono, T.; Fukushi, J.; Otsubo, M.; Onoue, H.; Ito, Y.; Inamura, T.; Ikezaki, K.; Fukui, M.; et al. Macrophage infiltration and heme oxygenase-1 expression correlate with angiogenesis in human gliomas. Clin. Cancer Res. 1999, 5, 1107–1113. [Google Scholar]
- Komohara, Y.; Ohnishi, K.; Kuratsu, J.; Takeya, M. Possible involvement of the M2 anti-inflammatory macrophage phenotype in growth of human gliomas. J. Pathol. 2008, 216, 15–24. [Google Scholar] [CrossRef]
- Markovic, D.S.; Glass, R.; Synowitz, M.; Rooijen, N.; Kettenmann, H. Microglia stimulate the invasiveness of glioma cells by increasing the activity of metalloprotease-2. J. Neuropathol. Exp. Neurol. 2005, 64, 754–762. [Google Scholar] [CrossRef]
- Markovic, D.S.; Vinnakota, K.; Chirasani, S.; Synowitz, M.; Raguet, H.; Stock, K.; Sliwa, M.; Lehmann, S.; Kalin, R.; van Rooijen, N.; et al. Gliomas induce and exploit microglial MT1-MMP expression for tumor expansion. Proc. Natl. Acad. Sci. USA 2009, 106, 12530–12535. [Google Scholar]
- Kioi, M.; Vogel, H.; Schultz, G.; Hoffman, R.M.; Harsh, G.R.; Brown, J.M. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J. Clin. Invest. 2010, 120, 694–705. [Google Scholar] [CrossRef]
- Pucci, F.; Venneri, M.A.; Biziato, D.; Nonis, A.; Moi, D.; Sica, A.; di Serio, C.; Naldini, L.; de Palma, M. A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophages suggests common functions and developmental relationships. Blood 2009, 114, 901–914. [Google Scholar]
- Galarneau, H.; Villeneuve, J.; Gowing, G.; Julien, J.P.; Vallieres, L. Increased glioma growth in mice depleted of macrophages. Cancer Res. 2007, 67, 8874–8881. [Google Scholar] [CrossRef]
- Kanamori, M.; Kawaguchi, T.; Berger, M.S.; Pieper, R.O. Intracranial microenvironment reveals independent opposing functions of host alphaVbeta3 expression on glioma growth and angiogenesis. J. Biol. Chem. 2006, 281, 37256–37264. [Google Scholar]
- Murata, J.; Ricciardi-Castagnoli, P.; Dessous L'Eglise Mange, P.; Martin, F.; Juillerat-Jeanneret, L. Microglial cells induce cytotoxic effects toward colon carcinoma cells: Measurement of tumor cytotoxicity with a gamma-glutamyl transpeptidase assay. Int. J. Cancer 1997, 70, 169–174. [Google Scholar] [CrossRef]
- Carson, M.J.; Doose, J.M.; Melchior, B.; Schmid, C.D.; Ploix, C.C. CNS immune privilege: Hiding in plain sight. Immunol. Rev. 2006, 213, 48–65. [Google Scholar] [CrossRef]
- Flugel, A.; Labeur, M.S.; Grasbon-Frodl, E.M.; Kreutzberg, G.W.; Graeber, M.B. Microglia only weakly present glioma antigen to cytotoxic T cells. Int. J. Dev. Neurosci. 1999, 17, 547–556. [Google Scholar] [CrossRef]
- Huettner, C.; Czub, S.; Kerkau, S.; Roggendorf, W.; Tonn, J.C. Interleukin 10 is expressed in human gliomas in vivo and increases glioma cell proliferation and motility in vitro. Anticancer Res. 1997, 17, 3217–3224. [Google Scholar]
- Kiefer, R.; Supler, M.L.; Toyka, K.V.; Streit, W.J. In situ detection of transforming growth factor-beta mRNA in experimental rat glioma and reactive glial cells. Neurosci. Lett. 1994, 166, 161–164. [Google Scholar] [CrossRef]
- Kostianovsky, A.M.; Maier, L.M.; Anderson, R.C.; Bruce, J.N.; Anderson, D.E. Astrocytic regulation of human monocytic/microglial activation. J. Immunol. 2008, 181, 5425–5432. [Google Scholar]
- Kuppner, M.C.; Sawamura, Y.; Hamou, M.F.; de Tribolet, N. Influence of PGE2- and cAMP-modulating agents on human glioblastoma cell killing by interleukin-2-activated lymphocytes. J. Neurosurg. 1990, 72, 619–625. [Google Scholar] [CrossRef]
- O'Keefe, G.M.; Nguyen, V.T.; Benveniste, E.N. Class II transactivator and class II MHC gene expression in microglia: Modulation by the cytokines TGF-beta, IL-4, IL-13 and IL-10. Eur. J. Immunol. 1999, 29, 1275–1285. [Google Scholar] [CrossRef]
- Wagner, S.; Czub, S.; Greif, M.; Vince, G.H.; Suss, N.; Kerkau, S.; Rieckmann, P.; Roggendorf, W.; Roosen, K.; Tonn, J.C. Microglial/macrophage expression of interleukin 10 in human glioblastomas. Int. J. Cancer 1999, 82, 12–16. [Google Scholar] [CrossRef]
- Schartner, J.M.; Hagar, A.R.; van Handel, M.; Zhang, L.; Nadkarni, N.; Badie, B. Impaired capacity for upregulation of MHC class II in tumor-associated microglia. Glia 2005, 51, 279–285. [Google Scholar] [CrossRef]
- Taniguchi, Y.; Ono, K.; Yoshida, S.; Tanaka, R. Antigen-presenting capability of glial cells under glioma-harboring conditions and the effect of glioma-derived factors on antigen presentation. J. Neuroimmunol. 2000, 111, 177–185. [Google Scholar] [CrossRef]
- Ogden, A.T.; Horgan, D.; Waziri, A.; Anderson, D.; Louca, J.; McKhann, G.M.; Sisti, M.B.; Parsa, A.T.; Bruce, J.N. Defective receptor expression and dendritic cell differentiation of monocytes in glioblastomas. Neurosurgery 2006, 59, 902–910. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
- Greten, T.F.; Manns, M.P.; Korangy, F. Myeloid derived suppressor cells in human diseases. Int. Immunopharmacol. 2011, 11, 802–807. [Google Scholar] [CrossRef]
- Talmadge, J.E. Pathways mediating the expansion and immunosuppressive activity of myeloid-derived suppressor cells and their relevance to cancer therapy. Clin. Cancer Res. 2007, 13, 5243–5248. [Google Scholar] [CrossRef]
- Melani, C.; Chiodoni, C.; Forni, G.; Colombo, M.P. Myeloid cell expansion elicited by the progression of spontaneous mammary carcinomas in c-erbB-2 transgenic BALB/c mice suppresses immune reactivity. Blood 2003, 102, 2138–2145. [Google Scholar] [CrossRef]
- Umemura, N.; Saio, M.; Suwa, T.; Kitoh, Y.; Bai, J.; Nonaka, K.; Ouyang, G.F.; Okada, M.; Balazs, M.; Adany, R.; et al. Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/macrophages that bear M1- and M2-type characteristics. J. Leukoc. Biol. 2008, 83, 1136–1144. [Google Scholar] [CrossRef]
- Gallina, G.; Dolcetti, L.; Serafini, P.; de Santo, C.; Marigo, I.; Colombo, M.P.; Basso, G.; Brombacher, F.; Borrello, I.; Zanovello, P.; et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J. Clin. Invest. 2006, 116, 2777–2790. [Google Scholar]
- Sofroniew, M.V. Reactive astrocytes in neural repair and protection. Neuroscientist 2005, 11, 400–407. [Google Scholar] [CrossRef]
- Sofroniew, M.V.; Vinters, H.V. Astrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 7–35. [Google Scholar] [CrossRef]
- Fontana, A.; Fierz, W.; Wekerle, H. Astrocytes present myelin basic protein to encephalitogenic T-cell lines. Nature 1984, 307, 273–276. [Google Scholar]
- Yong, V.W.; Yong, F.P.; Ruijs, T.C.; Antel, J.P.; Kim, S.U. Expression and modulation of HLA-DR on cultured human adult astrocytes. J. Neuropathol. Exp. Neurol. 1991, 50, 16–28. [Google Scholar] [CrossRef]
- Zhang, M.; Olsson, Y. Hematogenous metastases of the human brain—Characteristics of peritumoral brain changes: A review. J. Neurooncol. 1997, 35, 81–89. [Google Scholar] [CrossRef]
- Sierra, A.; Price, J. E.; Garcia-Ramirez, M.; Mendez, O.; Lopez, L.; Fabra, A. Astrocyte-derived cytokines contribute to the metastatic brain specificity of breast cancer cells. Lab. Invest. 1997, 77, 357–368. [Google Scholar]
- Seike, T.; Fujita, K.; Yamakawa, Y.; Kido, M.A.; Takiguchi, S.; Teramoto, N.; Iguchi, H.; Noda, M. Interaction between lung cancer cells and astrocytes via specific inflammatory cytokines in the microenvironment of brain metastasis. Clin. Exp. Metastasis 2010, 28, 13–25. [Google Scholar]
- Langley, R.R.; Fan, D.; Guo, L.; Zhang, C.; Lin, Q.; Brantley, E.C.; McCarty, J.H.; Fidler, I.J. Generation of an immortalized astrocyte cell line from H-2Kb-tsA58 mice to study the role of astrocytes in brain metastasis. Int. J. Oncol. 2009, 35, 665–672. [Google Scholar]
- Marchetti, D.; Li, J.; Shen, R. Astrocytes contribute to the brain-metastatic specificity of melanoma cells by producing heparanase. Cancer Res. 2000, 60, 4767–4770. [Google Scholar]
- Yoshida, K.; Gage, F.H. Fibroblast growth factors stimulate nerve growth factor synthesis and secretion by astrocytes. Brain Res. 1991, 538, 118–126. [Google Scholar] [CrossRef]
- Le, D.M.; Besson, A.; Fogg, D.K.; Choi, K.S.; Waisman, D.M.; Goodyer, C.G.; Rewcastle, B.; Yong, V.W. Exploitation of astrocytes by glioma cells to facilitate invasiveness: A mechanism involving matrix metalloproteinase-2 and the urokinase-type plasminogen activator-plasmin cascade. J. Neurosci. 2003, 23, 4034–4043. [Google Scholar]
- Lin, Q.; Balasubramanian, K.; Fan, D.; Kim, S.J.; Guo, L.; Wang, H.; Bar-Eli, M.; Aldape, K.D.; Fidler, I.J. Reactive astrocytes protect melanoma cells from chemotherapy by sequestering intracellular calcium through gap junction communication channels. Neoplasia 2010, 12, 748–754. [Google Scholar]
- Bechmann, I.; Steiner, B.; Gimsa, U.; Mor, G.; Wolf, S.; Beyer, M.; Nitsch, R.; Zipp, F. Astrocyte-induced T cell elimination is CD95 ligand dependent. J. Neuroimmunol. 2002, 132, 60–65. [Google Scholar]
- Jain, R.K.; di Tomaso, E.; Duda, D.G.; Loeffler, J.S.; Sorensen, A.G.; Batchelor, T.T. Angiogenesis in brain tumours. Nat. Rev. Neurosci. 2007, 8, 610–622. [Google Scholar]
- Batchelor, T.T.; Sorensen, A.G.; di Tomaso, E.; Zhang, W.T.; Duda, D.G.; Cohen, K.S.; Kozak, K.R.; Cahill, D.P.; Chen, P.J.; Zhu, M.; et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 2007, 11, 83–95. [Google Scholar] [CrossRef]
- JuanYin, J.; Tracy, K.; Zhang, L.; Munasinghe, J.; Shapiro, E.; Koretsky, A.; Kelly, K. Noninvasive imaging of the functional effects of anti-VEGF therapy on tumor cell extravasation and regional blood volume in an experimental brain metastasis model. Clin. Exp. Metastasis 2009, 26, 403–414. [Google Scholar] [CrossRef]
- Kim, L.S.; Huang, S.; Lu, W.; Lev, D.C.; Price, J.E. Vascular endothelial growth factor expression promotes the growth of breast cancer brain metastases in nude mice. Clin. Exp. Metastasis 2004, 21, 107–118. [Google Scholar] [CrossRef]
- Narayana, A.; Kelly, P.; Golfinos, J.; Parker, E.; Johnson, G.; Knopp, E.; Zagzag, D.; Fischer, I.; Raza, S.; Medabalmi, P.; et al. Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: Impact on local control and patient survival. J. Neurosurg. 2009, 110, 173–180. [Google Scholar]
- Norden, A.D.; Young, G.S.; Setayesh, K.; Muzikansky, A.; Klufas, R.; Ross, G.L.; Ciampa, A.S.; Ebbeling, L.G.; Levy, B.; Drappatz, J.; et al. Bevacizumab for recurrent malignant gliomas: Efficacy, toxicity, and patterns of recurrence. Neurology 2008, 70, 779–787. [Google Scholar]
- Folkins, C.; Man, S.; Xu, P.; Shaked, Y.; Hicklin, D.J.; Kerbel, R.S. Anticancer therapies combining antiangiogenic and tumor cell cytotoxic effects reduce the tumor stem-like cell fraction in glioma xenograft tumors. Cancer Res. 2007, 67, 3560–3564. [Google Scholar]
- de Palma, M.; Mazzieri, R.; Politi, L.S.; Pucci, F.; Zonari, E.; Sitia, G.; Mazzoleni, S.; Moi, D.; Venneri, M.A.; Indraccolo, S.; et al. Tumor-targeted interferon-alpha delivery by Tie2-expressing monocytes inhibits tumor growth and metastasis. Cancer Cell 2008, 14, 299–311. [Google Scholar] [CrossRef]
- Aboody, K.S.; Brown, A.; Rainov, N.G.; Bower, K.A.; Liu, S.; Yang, W.; Small, J.E.; Herrlinger, U.; Ourednik, V.; Black, P.M.; et al. Neural stem cells display extensive tropism for pathology in adult brain: Evidence from intracranial gliomas. Proc. Natl. Acad. Sci. USA 2000, 97, 12846–12851. [Google Scholar]
- Kim, S.K.; Kim, S.U.; Park, I.H.; Bang, J.H.; Aboody, K.S.; Wang, K.C.; Cho, B.K.; Kim, M.; Menon, L.G.; Black, P.M.; et al. Human neural stem cells target experimental intracranial medulloblastoma and deliver a therapeutic gene leading to tumor regression. Clin. Cancer Res. 2006, 12, 5550–5556. [Google Scholar]
- Bexell, D.; Gunnarsson, S.; Svensson, A.; Tormin, A.; Henriques-Oliveira, C.; Siesjo, P.; Paul, G.; Salford, L.G.; Scheding, S.; Bengzon, J. Rat multipotent mesenchymal stromal cells lack long-distance tropism to three different rat glioma models. Neurosurgery 2011. [Google Scholar]
- Balyasnikova, I.V.; Ferguson, S.D.; Sengupta, S.; Han, Y.; Lesniak, M.S. Mesenchymal stem cells modified with a single-chain antibody against EGFRvIII successfully inhibit the growth of human xenograft malignant glioma. PloS One 2010, 5, e9750. [Google Scholar]
© 2012 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/3.0/).
Share and Cite
Lorger, M. Tumor Microenvironment in the Brain. Cancers 2012, 4, 218-243. https://doi.org/10.3390/cancers4010218
Lorger M. Tumor Microenvironment in the Brain. Cancers. 2012; 4(1):218-243. https://doi.org/10.3390/cancers4010218
Chicago/Turabian StyleLorger, Mihaela. 2012. "Tumor Microenvironment in the Brain" Cancers 4, no. 1: 218-243. https://doi.org/10.3390/cancers4010218
APA StyleLorger, M. (2012). Tumor Microenvironment in the Brain. Cancers, 4(1), 218-243. https://doi.org/10.3390/cancers4010218