Review

Stem cell-based therapies for neurological disorders

  • Cell-based therapies have been previously performed using fetal tissues for some central nervous system (CNS) disorders, such as Parkinson’s disease. However, it can be difficult to collect a large number of cells for transplantation. Recent studies revealed that some stem cells can act as potential sources of cell-based therapies for degenerative and damaged areas in the CNS. In addition, stem cells can be used as cellular delivery vehicles for brain tumor because of tumor-tropic migratory capacity. Embryonic stem (ES) cells, mesenchymal stem cells (MSCs), and induced pluripotent stem (iPS) cells are the most attractive stem cells. iPS cells can be efficiently differentiated to neural stem cells and have the possibilities to overcome the ethical issues associated with ES cells. Therefore, cell-based therapies using iPS cells can be developed specifically for neurological disorders. In this article, we review the characteristics of ES cells, MSCs, and iPS cells as cell sources for stem cell-based therapies, and then discuss preclinical data and ongoing clinical trials for the CNS disorders.

    Citation: Ryota Tamura, Masahiro Toda. Stem cell-based therapies for neurological disorders[J]. AIMS Cell and Tissue Engineering, 2018, 2(1): 24-46. doi: 10.3934/celltissue.2018.1.24

    Related Papers:

  • Cell-based therapies have been previously performed using fetal tissues for some central nervous system (CNS) disorders, such as Parkinson’s disease. However, it can be difficult to collect a large number of cells for transplantation. Recent studies revealed that some stem cells can act as potential sources of cell-based therapies for degenerative and damaged areas in the CNS. In addition, stem cells can be used as cellular delivery vehicles for brain tumor because of tumor-tropic migratory capacity. Embryonic stem (ES) cells, mesenchymal stem cells (MSCs), and induced pluripotent stem (iPS) cells are the most attractive stem cells. iPS cells can be efficiently differentiated to neural stem cells and have the possibilities to overcome the ethical issues associated with ES cells. Therefore, cell-based therapies using iPS cells can be developed specifically for neurological disorders. In this article, we review the characteristics of ES cells, MSCs, and iPS cells as cell sources for stem cell-based therapies, and then discuss preclinical data and ongoing clinical trials for the CNS disorders.


    加载中
    [1] Okano H, Kaneko S, Okada S, et al. (2007) Regeneration-based therapies for spinal cord injuries. Neurochem Int 51: 68–73. doi: 10.1016/j.neuint.2007.04.013
    [2] Okano H, Sakaguchi M, Ohki K, et al. (2007) Regeneration of the central nervous system using endogenous repair mechanisms. J Neurochem 102: 1459–1465. doi: 10.1111/j.1471-4159.2007.04674.x
    [3] Okita K, Ichisaka T, Yamanaka S (2007) Generation of germ-line competent induced pluripotent stem cells. Nature 448: 313–317. doi: 10.1038/nature05934
    [4] Berker RA, Barrett J, Mason SL, et al. (2013) Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson's disease. Lancet Neurol 12: 84–91. doi: 10.1016/S1474-4422(12)70295-8
    [5] Horita Y, Honmou O, Harada K, et al. (2006) lntravenous administration of glial cell line derived neutrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. J Neurosci Res 84: 1495–1504. doi: 10.1002/jnr.21056
    [6] Morizane A, Doi D, Kikuchi T, et al. (2013) Direct comparison of autologous and allogeneic transplantation of iPSC-derived neural cells in the brain of a nonhuman primate. Stem Cell Reports 1: 283–292. doi: 10.1016/j.stemcr.2013.08.007
    [7] Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. (1998) Embryonic Stem Cell Lines Derived from Human. Science 282: 1145–1147.
    [8] Beyer Nardi N, da Silva Meirelles L (2006) Mesenchymal stem cells: isolation, in vitro expansion and characterization. Handb Exp Pharmacol 174: 249–282. doi: 10.1007/3-540-31265-X_11
    [9] Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126: 663–676. doi: 10.1016/j.cell.2006.07.024
    [10] Guzman R, Choi R, Gera A, et al. (2008) Intravascular cell replacement therapy for stroke. Neurosurg Focus 24: E15.
    [11] Matsushita T, Kibayashi T, Katayama T, et al. (2011) Mesenchymal stem cells transmigrate across brain microvascular endothelial cell monolayers through transiently formed inter-endothelial gaps. Neurosci Lett 502: 41–45. doi: 10.1016/j.neulet.2011.07.021
    [12] Steingen C, Brenig F, Baumgartner L, et al. (2008) Characterization of key mechanisms in transmigration and invasion of mesenchymal stem cells. J Mol Cell Cardiol 44: 1072–1084. doi: 10.1016/j.yjmcc.2008.03.010
    [13] Baldwing A (2009) Morality and human embryo research. Introduction to the Talking Point on morality and human embryo research. EMBO Reports 10: 299–300.
    [14] Henderson JK, Draper JS, Baillie HS, et al. (2002) Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. Stem Cells 20: 329–337. doi: 10.1634/stemcells.20-4-329
    [15] Hedlund E, Pruszak J, Lardaro T, et al. (2008) Embryonic stem cell-derived Pitx3-enhanced green fluorescent protein midbrain dopamine neurons survive enrichment by fluorescence-activated cell sorting and function in an animal model of Parkinson's disease. Stem Cells 26: 1526–1536. doi: 10.1634/stemcells.2007-0996
    [16] Dominici M, Le Blanc K, Mueller I, et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315–317.
    [17] Valero MC, Huntsman HD, Liu J, et al. (2012) Eccentric exercise facilitates mesenchymal stem cell appearance in skeletal muscle. PLoS One 7: e29760. doi: 10.1371/journal.pone.0029760
    [18] Wang S, Qu X, Zhao RC. (2012) Clinical applications of mesenchymal stem cells. J Hematol Oncol 5: 19. doi: 10.1186/1756-8722-5-19
    [19] Lalu MM, McIntyre L, Pugliese C, et al. (2012) Canadian Critical Care Trials Group. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One 7: e47559.
    [20] Mabuchi Y, Morikawa S, Harada S, et al. (2013) LNGFR(+)THY-1(+)VCAM-1(hi+) cells reveal functionally distinct subpopulations in mesenchymal stem cells. Stem Cell Reports 1: 152–165. doi: 10.1016/j.stemcr.2013.06.001
    [21] Morikawa S, Mabuchi Y, Kubota Y, et al. (2009) Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med 206: 2483–2496. doi: 10.1084/jem.20091046
    [22] Aggarwal S, Pittenger MF (2005) Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105: 1815–1822. doi: 10.1182/blood-2004-04-1559
    [23] Arinzeh TL, Peter SJ, Archambault MP, et al. (2003) Allogeneic mesenchymal stem cells regenerate bone in a critical-sized canine segmental defect. J Bone Joint Surg Am 85: 1927–1935. doi: 10.2106/00004623-200310000-00010
    [24] Farini A, Sitzia C, Erratico S, et al. (2014) Clinical applications of mesenchymal stem cells in chronic diseases. Stem Cells Int 2014: 306573.
    [25] Le Blanc K, Rasmusson I, Sundberg B, et al. (2004) Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 363: 1439–1441. doi: 10.1016/S0140-6736(04)16104-7
    [26] Tobin LM, Healy ME, English K, et al. (2013) Human mesenchymal stem cells suppress donor CD4(+) T cell proliferation and reduce pathology in a humanized mouse model of acute graft-versus-host disease. Clin Exp Immunol 172: 333–348. doi: 10.1111/cei.12056
    [27] Karussis D, Karageorgiou C, Vaknin-Dembinsky A, et al. (2010) Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol 67: 1187–1194.
    [28] von Bahr L, Batsis I, Moll G, et al. (2012) Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long-term engraftment and no ectopic tissue formation. Stem Cells 30: 1575–1578. doi: 10.1002/stem.1118
    [29] Takahashi K, Tanabe K, Ohnuki M, et al. (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861–872. doi: 10.1016/j.cell.2007.11.019
    [30] Johannesson B, Sagi I, Gore A, et al. (2014) Comparable frequencies of coding mutations and loss of imprinting in human pluripotent cells derived by nuclear transfer and defined factors. Cell Stem Cell 15: 634–642. doi: 10.1016/j.stem.2014.10.002
    [31] Ma H, Morey R, O'Neil RC, et al. (2014) Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature 511: 177–183. doi: 10.1038/nature13551
    [32] Okita K, Yamakawa T, Matsumura Y, et al. (2013) An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells 31: 458–466. doi: 10.1002/stem.1293
    [33] Warren L, Manos PD, Ahfeldt T, et al. (2010) Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7: 618–630. doi: 10.1016/j.stem.2010.08.012
    [34] McDonald JW, Liu XZ, Qu Y, et al. (1999) Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat Med 5: 1410–1412. doi: 10.1038/70986
    [35] Yu J, Vodyanik MA, Smuga-Otto K, et al. (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318: 1917–1920. doi: 10.1126/science.1151526
    [36] Hayashi Y, Chan T, Warashina M, et al. (2010) Reduction of N-glycolylneuraminic acid in human induced pluripotent stem cells generated or cultured under feeder-and serum-free defined conditions. PLoS One 5: e14099. doi: 10.1371/journal.pone.0014099
    [37] Miyazaki T, Futaki S, Suemori H, et al. (2012) Laminin E8 fragments support efficient adhesion and expansion of dissociated human pluripotent stem cells. Nat Commun 3: 1236. doi: 10.1038/ncomms2231
    [38] Rodin S, Domogatskaya A, Ström S, et al. (2010) Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nat Biotechnol 28: 611–615. doi: 10.1038/nbt.1620
    [39] Morita T, Sasaki M, Kataoka-Sasaki Y, et al. (2016) Intravenous infusion of mesenchymal stem cells promotes functional recovery in a model of chronic spinal cord injury. Neuroscience 335: 221–231. doi: 10.1016/j.neuroscience.2016.08.037
    [40] Nakajima F, Tokunaga K, Nakatsuji N (2007) Human leukocyte antigen matching estimations in a hypothetical bank of human embryonic stem cell lines in the Japanese population for use in cell transplantation therapy. Stem Cells 25: 983–985. doi: 10.1634/stemcells.2006-0566
    [41] Guo XL, Chen JS (2015) Research on induced pluripotent stem cells and the application in ocular tissues. Int J Ophthalmol 8: 818–825.
    [42] Chambers SM, Fasano CA, Papapetrou EP, et al. (2009) Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol 27: 275–280. doi: 10.1038/nbt.1529
    [43] Doi D, Morizane A, Kikuchi T, et al. (2012) Prolonged maturation culture favors a reduction in the tumorigencity and the dopaminergic function of human ESC-derived neural cells in a primate model of Parkinson's disease. Stem Cells 30: 935–945. doi: 10.1002/stem.1060
    [44] Kikuchi T, Morizane A, Doi D, et al. (2011) Survival of human induced pluripotent stem cell-derived midbrain dopaminergic neurons in the brain of a primate model of parkinson's disease. J Parkinsons Dis 1: 395–412.
    [45] Kriks S, Shim JW, Piano J, et al. (2011) Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. Nature 480: 547–551. doi: 10.1038/nature10648
    [46] Brederlau A, Correia AS, Anisimov SV, et al. (2006) Transplantation of human embryonic stem cell-derived cells to a rat model of Parkinson's disease: effect of in vitro differentiation on graft survival and teratoma formation. Stem Cells 24: 1433–1440. doi: 10.1634/stemcells.2005-0393
    [47] Grealish S, Diguet E, Kirkeby A, et al. (2014) Human EsC-derived dopamine neurons show similar preclinical efficacy and potency to fetal neurons wihen grafted in a rat model of Parkinson's disease. Cell stem Cell 15: 653–665. doi: 10.1016/j.stem.2014.09.017
    [48] Hayashi T, Wakao S, Kitada M, et al. (2013) Autologous mesenchymal stem cell-derived dopaminergic neurons function in parkinsonian macaques. J Clin Invest 123: 272–284. doi: 10.1172/JCI62516
    [49] Argus G, Cooper O, Deleidi M, et al. (2010) Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc Natl Acad Sci 107: 15921–15926. doi: 10.1073/pnas.1010209107
    [50] Rhee YH,Ko JY, Chang MY, et al. (2011) Protein-based human iPS cells efficient1y generate functional dopamine neurons and can treata rat model of Parkinson disease. J Clin Invest 121: 2326–2335. doi: 10.1172/JCI45794
    [51] Kikuchi T, Morizane A, Doi D, et al. (2017) Human iPS cell-derived dopaminergic neurons function in a primate Parkinson's disease model. Nature 548: 592–596. doi: 10.1038/nature23664
    [52] Barker RA, Drouin-Quellet J, Parmar M (2015) Cell-based therapies for Parkinson disease-past insights and future potential. Nat Rev Neurol 11: 492–503. doi: 10.1038/nrneurol.2015.123
    [53] Yan Liu, Jason P Weick, Huisheng Liu, et al. (2013) Medial ganglionic eminence–like cells derived from human embryonic stem cells correct learning and memory deficits. Nat Biotechnol 31: 440–447. doi: 10.1038/nbt.2565
    [54] Lee H, Shamy GA, Elkabetz Y, et al. (2007) Directed differentiation and transplantation of human embryonic stem cell-derived motoneurons. Stem Cells 25: 1931–1939. doi: 10.1634/stemcells.2007-0097
    [55] Lee JK, Jin HK, Bae JS (2010) Bone marrow-derived mesenchymal stem cells attenuate amyloid beta-induced memori impairment and apoptosis by inhibiting neuronal cell death. Curr Alzheimer Res 7: 540–548. doi: 10.2174/156720510792231739
    [56] Takamatsu K, Ikeda T, Haruta M, et al. (2014) Degradation of amyloid beta by human induced pluripotent stem cell-derived macrophages expressing Neprilysin-2. Stem Cell Res 13: 442–453. doi: 10.1016/j.scr.2014.10.001
    [57] Lee HJ, Lee JK, Lee H, et al. (2010) The therapeutic potential of human umbilical cord blood-derived mesenchymal stem cells in Alzheimer's disease. Neurosci Lett 481: 30–35. doi: 10.1016/j.neulet.2010.06.045
    [58] Harper JM, Krishnan C, Darman JS, et al. (2004) Axonal growth of embryonic stem cell-derived motoneurons in vitro and in motoneuron-injured adult rats. Proc Natl Acad Sci U S A 101: 7123–7128. doi: 10.1073/pnas.0401103101
    [59] Kerr DA, Lladó J, Shamblott MJ, et al. (2003) Human embryonic germ cell derivatives facilitate motor recovery of rats with diffuse motor neuron injury. J Neurosci 23: 5131–5140.
    [60] Lepore AC, Rauck B, Dejea C, et al. (2008) Focal transplantation-based astrocyte replacement is neuroprotective in a model of motor neuron disease. Nat Neurosci 11: 1294–1301. doi: 10.1038/nn.2210
    [61] Kawada J, Kaneda S, Kirihara T, et al. (2017) Generation of a Motor Nerve Organoid with Human Stem Cell-Derived Neurons. Stem Cell Reports 14: 1441–1449.
    [62] Aubry L, Bugi A, Lefort N, et al. (2008) Striatal progenitors derived from human ES cells mature into DARPP32 neurons in vitro and in quinolinic acid-lesioned rats. Proc Natl Acad Sci U S A 105: 16707–16712. doi: 10.1073/pnas.0808488105
    [63] Lee ST, Chu K, Jung KH, et al. (2009) Slowed progression in models of Huntington disease by adipose stem cell transplantation. Ann Neurol 66: 671–681. doi: 10.1002/ana.21788
    [64] Chae JI, Kim DW, Lee N, et al. (2012) Quantitative proteomic analysis of induced pluripotent stem cells derived from a human Huntington's disease patient. Biochem J 446: 359–371. doi: 10.1042/BJ20111495
    [65] Cicchetti F, Saporta S, Hauser RA, et al. (2009) Neural transplants in patients with Huntington's disease undergo disease-like neuronal degeneration. Proc Natl Acad Sci U S A 106: 12483–12488. doi: 10.1073/pnas.0904239106
    [66] Freeman TB, Cicchetti F, Hauser RA, et al. (2000) Transplanted fetal striatum in Huntington's disease:phenotypic development and lack of pathology. Proc Natl Acad Sci U S A 97: 13877–13882. doi: 10.1073/pnas.97.25.13877
    [67] Keene CD, Chang RC, Leverenz JB, et al. (2009) A patient with Huntington's disease and long-surviving fetal neural transplants that developed mass lesions. Acta Neuropathol 117: 329–338. doi: 10.1007/s00401-008-0465-0
    [68] Takagi Y, Hashimoto N (2006) Transplantation of embryonic stem cell-derived neural progenitors into ischemic brain. Nosotchu 28: 600–605. doi: 10.3995/jstroke.28.600
    [69] Hayashi J, Takagi Y, Fukuda H, et al. (2006) Primate embryonic stem cell-derived neuronal progenitors transplanted into ischemic brain. J Cereb Blood Flow Metab 26: 906–914. doi: 10.1038/sj.jcbfm.9600247
    [70] Chen S J, Chang CM, Tsai SK, et al. (2010) Functional improvementof focal cerebral ischemia injury by subdural transplantationof induced pluripotent stem cells with fibrin glue. Stem Cells Dev 19: 1757–1767. doi: 10.1089/scd.2009.0452
    [71] Suzuki J, Sasaki M, Harada K, et al. (2013) Bilateral cortical hyperactivity detencted by fMRI associates with improved motor function following intravenous infusion of mesenchymal stem cells in a rat stroke model. Brain Res 1497: 15–22. doi: 10.1016/j.brainres.2012.12.028
    [72] García R, Aguiar J, Alberti E, et al. (2004) Bone marrow stromal cells produce nerve growth factor and glial cell line-derived neurotrophic factors. Biochem Biophys Res Commun 316: 753–754. doi: 10.1016/j.bbrc.2004.02.111
    [73] Nomura T, Honmou O, Harada H, et al. (2005) Infusion of BDNF gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Neuroscience 136: 161–169. doi: 10.1016/j.neuroscience.2005.06.062
    [74] Onda T, Honmou O, Harada K, et al. (2008) Therapeutic benefits by human mesenchymal stem cells(hMSs) and Ang-1 gene-modifeied hMSCs after cerebral ischemia. J Cereb Blood Flow Metab 28: 329–340. doi: 10.1038/sj.jcbfm.9600527
    [75] Savitz SI, Chopp M, Deans R, et al. (2011) STEPS Participants: Stem Cell Therapy as an Emerging Paradigm for Stroke (STEPS) II. Stroke 42: 825–829. doi: 10.1161/STROKEAHA.110.601914
    [76] Uemura M, Kasahara Y, Nagatsuka K, et al. (2012) Cell-based therapy to promote angiogenesis in the brain following ischemic damage. Curr Vasc Pharmacol 10: 285–288. doi: 10.2174/157016112799959369
    [77] Hermanto Y, Sunohara T, Faried A, et al. (2017) Transplantation of feeder-free human induced pluripotent stem cell-derived cortical neuron progenitors in adult male Wistar rats with focal brain ischemia. J Neurosci Res 96: 863–874.
    [78] Honmou O, Houkin K, Matsunaga T, et al. (2011) Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain 134: 1790–1807. doi: 10.1093/brain/awr063
    [79] Steinberg GK, Kondziolka D, Wechsler LR, et al. (2016) Clinical Outcomes of Transplanted Modified Bone Marrow-Derived Mesenchymal Stem Cells in Stroke: A Phase 1/2a Study. Stroke 47: 1817–1824. doi: 10.1161/STROKEAHA.116.012995
    [80] Kumagai G, Okada Y, Yamane J, et al. (2009) Roles of ES cell-derived gliogenic neural stem/progenitor cells in functional recovery after spinal cord injury. PLoS One 4: e7706. doi: 10.1371/journal.pone.0007706
    [81] Erceg S, Ronaghi M, Oria M, et al. (2010) Transplanted oligodendrocytes and motoneuron progenitors generated from human embryonic stem cells promote locomotor recovery after spinal cord transaction. Stem Cells 28: 1541–1549. doi: 10.1002/stem.489
    [82] Keirstead HS, Nistor G, Bernal G, et al. (2005) Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci 25: 4694–4705. doi: 10.1523/JNEUROSCI.0311-05.2005
    [83] Osaka M, Honmou O, Murakami T, et al. (2010) Intravenous administration of mesenchymal stem cells derived from bone marrow after contusive spinal cord injury improves functional outcome. Brain Res 9: 226–235.
    [84] Müller FJ, Snyder EY, Loring JF (2006) Gene therapy: can neural stem cells deliver? Nat Rev Neurosci 7: 75–84. doi: 10.1038/nrn1829
    [85] Tsuji O, Miura K, Okada Y, et al. (2010) Therapeutic potential of appropriately evaluated safe-induced pluripotent stem cells for spinal cord injury. Proc Natl Acad Sci U S A 107: 12704–12709. doi: 10.1073/pnas.0910106107
    [86] Fujimoto Y, Abematsu M, Falk A, et al. (2012) Treatment of a mouse model of spinal cord injury by transplantation of human induced pluripotent stem cell-derived long-term self-renewing neuroepithelial-like stem cells. Stem Cells 30: 1163–1173. doi: 10.1002/stem.1083
    [87] Nori S, Okada Y, Yasuda A, et al. (2011) Grafted human-induced pluripotent stem-cell-derived neurospheres promote motor functional recovery after spinal cord injury in mice. Proc Natl Acad Sci U S A 108: 16825–16830.
    [88] Kobayashi Y, Okada Y, Itakura G, et al. (2012) Pre-evaluated safe human iPSC-derived neural stem cells promote functional recovery after spinal cord injury in common marmoset without tumorigenicity. PLoS One 7: e52787. doi: 10.1371/journal.pone.0052787
    [89] Nishimura S, Yasuda A, Iwai H, et al. (2013) Time-dependent changes in the microenvironment of injured spinal cord affects the therapeutic potential of neural stem cell transplantation for spinal cord injury. Mol Brain 6: 3. doi: 10.1186/1756-6606-6-3
    [90] Kuroda Y, Kitada M, Wakao S, et al. (2011) Bone marrow mesenchymal cells: how do they contribute to tissue repair and are they really stem cells? Arch lmmunol Ther Exp 59: 369–378. doi: 10.1007/s00005-011-0139-9
    [91] Yoon SH, Shim YS, Park YH, et al. (2007) Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage-colony stimulating factor: Phase I/II clinical trial. Stem Cells 25: 2066–2073. doi: 10.1634/stemcells.2006-0807
    [92] Riess P, Molcanyi M, Bentz K, et al. (2007) Embryonic stem cell transplantation after experimental traumatic brain injury dramatically improves neurological outcome, but may cause tumors. J Neurotrauma 24: 216–225. doi: 10.1089/neu.2006.0141
    [93] Tajiri N, Duncan K, Antoine A, et al. (2014) Stem cell-paved biobridge facilitates neural repair in traumatic brain injury. Front Syst Neurosci 8: 116.
    [94] Tian C, Wang X, Wang X, et al. (2013) Autologous bone marrow mesenchymal stem cell therapy in the subacute stage of traumatic brain injury by lumbar puncture. Exp Clin Transplant 11: 176–181. doi: 10.6002/ect.2012.0053
    [95] Stupp R, Mason WP, van den Bent MJ, et al. (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352: 987–996. doi: 10.1056/NEJMoa043330
    [96] Stupp R, Brada M, van den Bent MJ, et al. (2014) ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25: 93–101.
    [97] Kim SU (2011) Neural stem cell-based gene therapy for brain tumors. Stem Cell Rev 7: 130–140. doi: 10.1007/s12015-010-9154-1
    [98] Rainov N (2000) A phase III clinical evaluation of herpes simplex virus type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum Gene Ther 11: 2389401.
    [99] Nakamura K, Ito Y, Kawano Y, et al. (2004) Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther 11: 1155–1164. doi: 10.1038/sj.gt.3302276
    [100] Stagg J, Lejeune L, Paquin A, et al. (2004) Marrow stromal cells for interleukin-2 delivery in cancer immunotherapy. Hum Gene Ther 15: 597–608. doi: 10.1089/104303404323142042
    [101] Yuan X, Hu J, Belladonna ML, et al. (2006) Interleukin-23-expressing bone marrow-derived neural stem-like cells exhibit antitumor activity against intracranial glioma. Cancer Res 66: 2630–2638. doi: 10.1158/0008-5472.CAN-05-1682
    [102] Kim SM, Lim JY, Park SI, et al. (2008) Gene therapy using TRAIL-secreting human umbilical cord blood-derived mesenchymal stem cells against intracranial glioma. Cancer Res 68: 9614–9623. doi: 10.1158/0008-5472.CAN-08-0451
    [103] Ahmed AU, Rolle CE, Tyler MA, et al. (2010) Bone marrow mesenchymal stem cells loaded with an oncolytic adenovirus suppress the anti-adenoviral immune response in the cotton rat model. Mol Ther 18: 1846–1856. doi: 10.1038/mt.2010.131
    [104] Herrlinger U, Woiciechowski C, Sena-Esteves M, et al. (2000) Neural precursor cells for delivery of replication-conditional HSV-1 vectors to intracerebral gliomas. Mol Ther 1: 347–357. doi: 10.1006/mthe.2000.0046
    [105] Tyler MA, Ulasov IV, Sonabend AM, et al. (2009) Neural stem cells target intracranial glioma to deliver an oncolytic adenovirus in vivo. Gene Ther 16: 262–278. doi: 10.1038/gt.2008.165
    [106] Rigg A, Sikora K (1997) Genetic prodrug activation therapy. Mol Med Today 3: 359–366. doi: 10.1016/S1357-4310(97)01082-4
    [107] Kim JH, Kim JY, Kim SU, et al. (2012) Therapeutic effect of genetically modified human neural stem cells encoding cytosine deaminase on experimental glioma. Biochem Biophys Res Commun 417: 534–540. doi: 10.1016/j.bbrc.2011.11.155
    [108] Luo Y, Zhu D, Lam DH, et al. (2015) A Double-Switch Cell Fusion-Inducible Transgene Expression System for Neural Stem Cell-Based Antiglioma Gene Therapy. Stem Cells Int 2015: 649080.
    [109] Amano S, Li S, Gu C, et al. (2009) Use of genetically engineered bone marrow-derived mesenchymal stem cells for glioma gene therapy. Int J Oncol 35: 1265–1270.
    [110] Miletic H, Fischer YH, Litwak S, et al. (2007) Bystander killing of malignant glioma by bone marrow-derived tumor-infiltrating progenitor cells expressing a suicide gene. Mol Ther 15: 1373–1381. doi: 10.1038/sj.mt.6300155
    [111] Lee EX, Lam DH, Wu C, et al. (2011) Glioma gene therapy using induced pluripotent stem cell derived neural stem cells. Mol Pharm 8: 1515–1524. doi: 10.1021/mp200127u
  • Reader Comments
  • © 2018 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(4109) PDF downloads(1071) Cited by(1)

Article outline

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog