1,731
Views
46
CrossRef citations to date
0
Altmetric
Reviews

Gene therapy for neurological disorders

, &
Pages 143-159 | Received 17 Jul 2015, Accepted 26 Oct 2015, Published online: 05 Dec 2015

References

  • Papers of special note have been highlighted as
  • • of interest
  • Shimada M, Abe S, Takahashi T, et al. Prophylaxis and treatment of Alzheimer’s disease by delivery of an adeno-associated virus encoding a monoclonal antibody targeting the amyloid Beta protein. PLoS One. 2013;8:e57606.
  • Fukuchi K, Tahara K, Kim HD, et al. Anti-Abeta single-chain antibody delivery via adeno-associated virus for treatment of Alzheimer’s disease. Neurobiol Dis. 2006;23:502–511.
  • Ryan DA, Mastrangelo MA, Narrow WC, et al. Abeta-directed single-chain antibody delivery via a serotype-1 AAV vector improves learning behavior and pathology in Alzheimer’s disease mice. Mol Ther. 2010;18:1471–1481.
  • Kou J, Kim H, Pattanayak A, et al. Anti-amyloid-β single-chain antibody brain delivery via AAV reduces amyloid load but may increase cerebral hemorrhages in an Alzheimer’s disease mouse model. J Alzheimers Dis. 2011;27:23–38.
  • Levites Y, Jansen K, Smithson LA, et al. Intracranial adeno-associated virus-mediated delivery of anti-pan amyloid beta, amyloid beta40, and amyloid beta42 single-chain variable fragments attenuates plaque pathology in amyloid precursor protein mice. J Neurosci. 2006;26:11923–11928.
  • Zhang J, Wu X, Qin C, et al. A novel recombinant adeno-associated virus vaccine reduces behavioral impairment and beta-amyloid plaques in a mouse model of Alzheimer’s disease. Neurobiol Dis. 2003;14:365–379.
  • Mouri A, Noda Y, Hara H, et al. Oral vaccination with a viral vector containing Abeta cDNA attenuates age-related Abeta accumulation and memory deficits without causing inflammation in a mouse Alzheimer model. Faseb J. 2007;21:2135–2148.
  • Iwata N, Higuchi M, Saido TC. Metabolism of amyloid-beta peptide and Alzheimer’s disease. Pharmacol Ther. 2005;108:129–148.
  • Iwata N, Mizukami H, Shirotani K, et al. Presynaptic localization of neprilysin contributes to efficient clearance of amyloid-beta peptide in mouse brain. J Neurosci. 2004;24:991–998.
  • Iwata N, Sekiguchi M, Hattori Y, et al. Global brain delivery of neprilysin gene by intravascular administration of AAV vector in mice. Sci Rep. 2013;3:1472.
  • Liu Y, Studzinski C, Beckett T, et al. Expression of neprilysin in skeletal muscle reduces amyloid burden in a transgenic mouse model of Alzheimer disease. Mol Ther. 2009;17:1381–1386.
  • Carty NC, Nash K, Lee D, et al. Adeno-associated viral (AAV) serotype 5 vector mediated gene delivery of endothelin-converting enzyme reduces Abeta deposits in APP + PS1 transgenic mice. Mol Ther. 2008;16:1580–1586.
  • Pascual-Lucas M, Viana Da Silva S, Di Scala M, et al Insulin-like growth factor 2 reverses memory and synaptic deficits in APP transgenic mice. EMBO Mol Med. 2014;6:1246–1262.
  • Tuszynski MH, Thal L, Pay M, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11:551–555.

• Efficacy of ex vivo nerve growth factor (NGF) gene delivery was demonstrated in patients with Alzheimer’s disease (AD).

  • Mandel RJ. CERE-110, an adeno-associated virus-based gene delivery vector expressing human nerve growth factor for the treatment of Alzheimer’s disease. Curr Opin Mol Ther. 2010;12:240–247.

• CERE-110 (AAV2-NGF) has passed phase I clinical testing and a multicenter phase II clinical trial has commenced.

  • Kiyota T, Okuyama S, Swan RJ, et al. CNS expression of anti-inflammatory cytokine interleukin-4 attenuates Alzheimer’s disease-like pathogenesis in APP+PS1 bigenic mice. Faseb J. 2010;24:3093–3102.
  • Kiyota T, Ingraham KL, Swan RJ, et al. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP+PS1 mice. Gene Ther. 2012;19:724–733.
  • Kiyota T, Zhang G, Morrison CM, et al. AAV2/1 CD74 gene transfer reduces β-amyloidosis and improves learning and memory in a mouse model of Alzheimer’s Disease. Mol Ther. 2015. [Epub ahead of print].
  • Murphy SR, Chang CC, Dogbevia G, et al. Acat1 knockdown gene therapy decreases amyloid-β in a mouse model of Alzheimer’s disease. Mol Ther. 2013;21:1497–1506.
  • Hudry E, Van Dam D, Kulik W, et al. Adeno-associated virus gene therapy with cholesterol 24-hydroxylase reduces the amyloid pathology before or after the onset of amyloid plaques in mouse models of Alzheimer’s disease. Mol Ther. 2010;18:44–53.
  • Eberling JL, Jagust WJ, Christine CW, et al. Results from a phase I safety trial of hAADC gene therapy for Parkinson disease. Neurology. 2008;70:1980–1983.
  • Christine CW, Starr PA, Larson PS, et al. Safety and tolerability of putaminal AADC gene therapy for Parkinson disease. Neurology. 2009;73:1662–1669.

• Class IV evidence that bilateral intrastriatal infusion of AAV2-hAADC improves mean unified Parkinson disease rating scale (UPDRS) score in Parkinson’s disease (PD) patients.

  • Muramatsu S, Fujimoto K, Kato S, et al. A phase I study of aromatic l-amino acid decarboxylase gene therapy for Parkinson’s disease. Mol Ther. 2010;18:1731–1735.

• Class IV evidence regarding the safety and efficacy of AADC gene therapy warrants further evaluation in a randomized, controlled, phase II setting.

  • Mittermeyer G, Christine CW, Rosenbluth KH, et al. Long-term evaluation of a phase 1 study of AADC gene therapy for Parkinson’s disease. Hum Gene Ther. 2012;23:377–381.

• Apparent efficacy of AAV2-AADC continues through later years with an acceptable safety profile.

• A phase I/II clinical trial involving ProSavin® delivery into the bilateral putamen reported improvement in motor function in all PD cohorts at primary efficacy end point.

  • Kaplitt MG, Feigin A, Tang C, et al. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson’s disease: an open label, phase I trial. Lancet. 2007;369:2097–2105.
  • LeWitt PA, Rezai AR, Leehey MA, et al. AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial. AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol. 2011;10:309–319.

• The efficacy and safety of bilateral infusion of AAV2-GAD in the subthalamic nucleus supports its further development for PD treatment.

  • Lang AE, Gill S, Patel NK, et al. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann Neurol. 2006;59:459–466.
  • Marks WJ Jr, Ostrem JL, Verhagen L, et al. Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson’s disease: an open-label, phase I trial. Lancet Neurol. 2008;7:400–408.
  • Marks WJ Jr, Bartus RT, Siffert J, et al. Gene delivery of AAV2-neurturin for Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neurol. 2010;9:1164–1172.

• A double-blind, phase II randomized trial in patients with advanced PD showed that intraputaminal AAV2-NTN is not superior to sham surgery at a primary end point of 12 months, but did report positive results in the subgroup of patients who were assessed for up to 18 months.

  • Wang YL, Liu W, Wada E, et al. Clinico-pathological rescue of a model mouse of Huntington’s disease by siRNA. Neurosci Res. 2005;53:241–249.
  • Harper SQ, Staber PD, He X, et al. RNA interference improves motor and neuropathological abnormalities in a Huntington’s disease mouse model. Proc Natl Acad Sci USA. 2005;19(102):5820–5825.
  • Rodriguez-Lebron E, Denovan-Wright EM, Nash K, et al. Intrastriatal rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington’s disease transgenic mice. Mol Ther. 2005;12:618–633.
  • Reiner A, Dragatsis I, Zeitlin S, et al. Wild-type huntingtin plays a role in brain development and neuronal survival. Mol Neurobiol. 2003;28:259–276.
  • Drouet V, Perrin V, Hassig R, et al. Sustained effects of nonallele-specific Huntingtin silencing. Ann Neurol. 2009;65:276–285.
  • Boudreau RL, McBride JL, Martins I, et al. Nonallele-specific silencing of mutant and wild-type huntingtin demonstrates therapeutic efficacy in Huntington’s disease mice. Mol Ther. 2009;17:1053–1063.
  • Kordasiewicz HB, Stanek LM, Wancewicz EV, et al. Sustained therapeutic reversal of Huntington’s disease by transient repression of huntingtin synthesis. Neuron. 2012;74:1031–1044.
  • McBride JL, Ramaswamy S, Gasmi M, et al. Viral delivery of glial cell line-derived neurotrophic factor improves behavior and protects striatal neurons in a mouse model of Huntington’s disease. Proc Natl Acad Sci U S A. 2006;103:9345–9350.
  • Ebert AD, Barber AE, Heins BM, et al. Ex vivo delivery of GDNF maintains motor function and prevents neuronal loss in a transgenic mouse model of Huntington’s disease. Exp Neurol. 2010 Jul;224(1):155–162.
  • Pérez-Navarro E, Akerud P, Marco S, et al. Neurturin protects striatal projection neurons but not interneurons in a rat model of Huntington’s disease. Neuroscience. 2000;98:89–96.
  • Ramaswamy S, McBride JL, Herzog CD, et al. Neurturin gene therapy improves motor function and prevents death of striatal neurons in a 3-nitropropionic acid rat model of Huntington’s disease. Neurobiol Dis. 2007;26:375–384.
  • Ramaswamy S, McBride JL, Han I, et al. Intrastriatal CERE-120 (AAV-Neurturin) protects striatal and cortical neurons and delays motor deficits in a transgenic mouse model of Huntington’s disease. Neurobiol Dis. 2009;34:40–50.
  • Zuccato C, Ciammola A, Rigamonti D, et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington’s disease. Science. 2001;293:493–498.
  • Kells AP, Henry RA, Connor B. AAV-BDNF mediated attenuation of quinolinic acid-induced neuropathology and motor function impairment. Gene Ther. 2008;15:966–977.
  • Bachoud-Lévi AC, Déglon N, Nguyen JP, et al. Neuroprotective gene therapy for Huntington’s disease using a polymer encapsulated BHK cell line engineered to secrete human CNTF. Hum Gene Ther. 2000;11:1723–1729.
  • Bloch J, Bachoud-Lévi AC, Déglon N, et al. Neuroprotective gene therapy for Huntington’s disease, using polymer-encapsulated cells engineered to secrete human ciliary neurotrophic factor: results of a phase I study. Hum Gene Ther. 2004;15:968–975.

• A phase I study in which polymer-encapsulated cells of a BHK cell line secreting ciliary neurotrophic factor (CNTF) were transplanted into the lateral ventricles of Huntington’s disease (HD) patients reported no sign of toxicity and also improvement in electrophysiological results.

  • Thomsen GM, Gowing G, Latter J, et al. Delayed Disease Onset and Extended Survival in the SOD1G93A Rat Model of Amyotrophic Lateral Sclerosis after Suppression of Mutant SOD1 in the Motor Cortex. J Neurosci. 2014;34:15587–15600.
  • Jackson KL, Dayton RD, Orchard EA, et al. Preservation of forelimb function by UPF1 gene therapy in a rat model of TDP-43-induced motor paralysis. Gene Ther. 2015;22:20–28.
  • Sareen D, O’Rourke JG, Meera P, et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci Transl Med. 2013;5:208ra149.
  • Lagier-Tourenne C, Baughn M, Rigo F, et al. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc Natl Acad Sci U S A. 2013;110:E4530–E4539.
  • Riboldi G, Zanetta C, Ranieri M, et al. Antisense Oligonucleotide Therapy for the Treatment of C9ORF72 ALS/FTD Diseases. Mol Neurobiol. 2014;50:721–732.
  • Manabe Y, Nagano I, Gazi MS, et al. Adenovirus-mediated gene transfer of glial cell line-derived neurotrophic factor prevents motor neuron loss of transgenic model mice for amyotrophic lateral sclerosis. Apoptosis. 2002;7:329–334.
  • Wang LJ, Lu YY, Muramatsu S, et al. Neuroprotective effects of glial cell line-derived neurotrophic factor mediated by an adeno-associated virus vector in a transgenic animal model of amyotrophic lateral sclerosis. J Neurosci. 2002;22:6920–6928.
  • Lu YY, Wang LJ, Muramatsu S, et al. Intramuscular injection of AAV-GDNF results in sustained expression of transgenic GDNF, and its delivery to spinal motoneurons by retrograde transport. Neurosci Res. 2003;45:33–40.
  • Guillot S, Azzouz M, Déglon N, et al. Local GDNF expression mediated by lentiviral vector protects facial nerve motoneurons but not spinal motoneurons in SOD1(G93A) transgenic mice. Neurobiol Dis. 2004;16:139–149.
  • Mohajeri MH, Figlewicz DA, Bohn MC. Intramuscular grafts of myoblasts genetically modified to secrete glial cell line-derived neurotrophic factor prevent motoneuron loss and disease progression in a mouse model of familial amyotrophic lateral sclerosis. Hum Gene Ther. 1999;10:1853–1866.
  • Suzuki M, McHugh J, Tork C, et al. Direct muscle delivery of GDNF with human mesenchymal stem cells improves motor neuron survival and function in a rat model of familial ALS. Mol Ther. 2008;16:2002–2010.
  • Kaspar BK, Lladó J, Sherkat N, et al. Retrograde viral delivery of IGF-1 prolongs survival in a mouse ALS model. Science. 2003;8(301):839–842.
  • Dodge JC, Haidet AM, Yang W, et al. Delivery of AAV-IGF-1 to the CNS extends survival in ALS mice through modification of aberrant glial cell activity. Mol Ther. 2008;16:1056–1064.
  • Dodge JC, Treleaven CM, Fidler JA, et al. AAV4-mediated expression of IGF-1 and VEGF within cellular components of the ventricular system improves survival outcome in familial ALS mice. Mol Ther. 2010;18:2075–2084.
  • Krakora D, Mulcrone P, Meyer M, et al. Synergistic effects of GDNF and VEGF on lifespan and disease progression in a familial ALS rat model. Mol Ther. 2013;21:1602–1610.
  • Borasio GD, Robberecht W, Leigh PN, et al. A placebo-controlled trial of insulin-like growth factor-I in amyotrophic lateral sclerosis. European ALS/IGF-I Study Group. Neurology. 1998;51:583–586.
  • Beauverd M, Mitchell JD, Wokke JH, et al. Recombinant human insulin-like growth factor I (rhIGF-I) for the treatment of amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2012;11:CD002064.
  • Xu DG, Crocker SJ, Doucet JP. Elevation of neuronal expression of NAIP reduces ischemic damage in the rat hippocampus. Nat Med. 1997;3:997–1004.
  • Yang GY, Pang L, Ge HL, et al. Attenuation of ischemia-induced mouse brain injury by SAG, a redox-inducible antioxidant protein. J Cereb Blood Flow Metab. 2001;21:722–733.
  • Shimazaki K, Urabe M, Monahan J, et al. Adeno-associated virus vector-mediated bcl-2 gene transfer into post-ischemic gerbil brain in vivo: prospects for gene therapy of ischemia-induced neuronal death. Gene Ther. 2000;7:1244–1249.
  • Okada T, Shimazaki K, Nomoto T, et al. Adeno-associated viral vector-mediated gene therapy of ischemia-induced neuronal death. Met Enzymol. 2002;346:378–393.
  • Sun Y, Jin K, Clark KR, et al. Adeno-associated virus-mediated delivery of BCL-w gene improves outcome after transient focal cerebral ischemia. Gene Ther. 2003;10:115–122.
  • Betz AL, Yang GY, Davidson BL. Attenuation of stroke size in rats using an adenoviral vector to induce overexpression of interleukin-1 receptor antagonist in brain. J Cereb Blood Flow Metab. 1995;15:547–551.
  • Works MG, Koenig JB, Sapolsky RM. Soluble TNF receptor 1-secreting ex vivo-derived dendritic cells reduce injury after stroke. J Cereb Blood Flow Metab. 2013 Sep;33(9):1376–1385.
  • Bellomo M, Adamo EB, Deodato B, et al. Enhancement of expression of vascular endothelial growth factor after adeno-associated virus gene transfer is associated with improvement of brain ischemia injury in the gerbil. Pharmacol Res. 2003;48:309–317.
  • Andsberg G, Kokaia Z, Klein RL, et al. Neuropathological and behavioral consequences of adeno-associated viral vector-mediated continuous intrastriatal neurotrophin delivery in a focal ischemia model in rats. Neurobiol Dis. 2002;9:187–204.
  • Gustafsson E, Andsberg G, Darsalia V, et al. Anterograde delivery of brain-derived neurotrophic factor to striatum via nigral transduction of recombinant adeno-associated virus increases neuronal death but promotes neurogenic response following stroke. Eur J Neurosci. 2003;17:2667–2678.
  • Larsson E, Mandel RJ, Klein RL, et al. Suppression of insult-induced neurogenesis in adult rat brain by brain-derived neurotrophic factor. Exp Neurol. 2002;177:1–8.
  • Tsai TH, Chen SL, Chiang YH, et al. Recombinant adeno-associated virus vector expressing glial cell line-derived neurotrophic factor reduces ischemia-induced damage. Exp Neurol. 2000;166:266–275.
  • Arvidsson A1, Kirik D, Lundberg C, et al. Elevated GDNF levels following viral vector-mediated gene transfer can increase neuronal death after stroke in rats. Neurobiol Dis. 2003;14:542–546.
  • Hu Q, Chen C, Khatibi NH, et al. Lentivirus-mediated transfer of MMP-9 shRNA provides neuroprotection following focal ischemic brain injury in rats. Brain Res. 2011;1367:347–359.
  • Schebesch KM, Herbst A, Bele S, et al. Calcitonin-gene related peptide and cerebral vasospasm. J Clin Neurosci. 2013;20:584–586.
  • Toyoda K, Faraci FM, Watanabe Y, et al. Gene transfer of calcitonin gene-related peptide prevents vasoconstriction after subarachnoid hemorrhage. Circ Res. 2000;87:818–824.
  • Stoodley M, Weihl CC, Zhang ZD, et al. Effect of adenovirus-mediated nitric oxide synthase gene transfer on vasospasm after experimental subarachnoid hemorrhage. Neurosurgery. 2000;46:1193–1202.
  • Onoue H, Tsutsui M, Smith L, et al. Expression and function of recombinant endothelial nitric oxide synthase gene in canine basilar artery after experimental subarachnoid hemorrhage. Stroke. 1998;29:1959–1965.
  • Santhanam AV, Smith LA, Akiyama M, et al. Role of endothelial NO synthase phosphorylation in cerebrovascular protective effect of recombinant erythropoietin during subarachnoid hemorrhage-induced cerebral vasospasm. Stroke. 2005;36:2731–2737.
  • Grieger JC, Samulski RJ. Adeno-associated virus as a gene therapy vector: vector development, production and clinical applications. Adv Biochem Eng Biotechnol. 2005;99:119–145.
  • Watakabe A, Ohtsuka M, Kinoshita M, et al. Comparative analyses of adeno-associated viral vector serotypes 1, 2, 5, 8 and 9 in marmoset, mouse and macaque cerebral cortex. Neurosci Res. 2015;93:144–157.
  • European Society of Gene Therapy. French gene therapy group reports on the adverse event in a clinical trial of gene therapy for X-linked severe combined immune deficiency (X-SCID). Position statement from the European Society of Gene Therapy. J Gene Med. 2003;5:82–84.
  • Wilson JM. Lessons learned from the gene therapy trial for ornithine transcarbamylase deficiency. Mol Genet Metab. 2009;96:151–157.
  • Prasad KN, Clarkson ED, La Rosa FG, et al. Efficacy of grafted immortalized dopamine neurons in an animal model of parkinsonism. Mol Genet Metab. 1998;65:1–9.
  • Rohrer DC, Nilaver G, Nipper V, et al. Genetically modified PC12 brain grafts: survivability and inducible nerve growth factor expression. Cell Transplant. 1996;5:57–68.
  • Fricker RA, Carpenter MK, Winkler C, et al. Site-specific migration and neuronal differentiation of human neural progenitor cells after transplantation in the adult rat brain. J Neurosci. 1999;15(19):5990–6005.
  • Yoshimoto Y, Lin Q, Collier TJ, et al. Astrocytes retrovirally transduced with BDNF elicit behavioral improvement in a rat model of Parkinson’s disease. Brain Res. 1995;691:25–36.
  • Ljungberg MC, Stern G, Wilkin GP. Survival of genetically engineered, adult-derived rat astrocytes grafted into the 6-hydroxydopamine lesioned adult rat striatum. Brain Res. 1999;816:29–37.
  • Bankiewicz KS, Forsayeth J, Eberling JL, et al. Long-term clinical improvement in MPTP-lesioned primates after gene therapy with AAV-hAACD. Mol Ther. 2006;14:564–570.
  • Fan DS, Ogawa M, Fujimoto KI, et al. Behavioral recovery in 6-hydroxydopamine-lesioned rats by cotransduction of striatum with tyrosine hydroxylase and aromatic l-amino acid decarboxylase genes using two separate adeno-associated virus vectors. Hum Gene Ther. 1998;9:2527–2535.
  • During MJ, Samulski RJ, Elsworth JD, et al. In vivo expression of therapeutic human genes for dopamine production in the caudates of MPTP-treated monkeys using an AAV vector. Gene Ther. 1998;5:820–827.
  • Azzouz M, Martin-Rendon E, Barber RD, et al. Multicistronic lentiviral vector-mediated striatal gene transfer of aromatic l-amino acid decarboxylase, tyrosine hydroxylase, and GTP cyclohydrolase I induces sustained transgene expression, dopamine production, and functional improvement in a rat model of Parkinson’s disease. J Neurosci. 2002;22:10302–10312.
  • Jarraya B, Boulet S, Ralph GS, et al. Dopamine gene therapy for Parkinson’s disease in a nonhuman primate without associated dyskinesia. Sci Transl Med. 2009;1:2ra4.
  • Luo J, Kaplitt MG, Fitzsimons HL, et al. Subthalamic GAD gene therapy in a Parkinson’s disease rat model. Science. 2002;298:425–429.
  • Emborg ME, Carbon M, Holden JE, et al. Subthalamic glutamic acid decarboxylase gene therapy: changes in motor function and cortical metabolism. J Cereb Blood Flow Metab. 2007;27:501–509.
  • Wang L, Muramatsu S, Lu Y, et al. Delayed delivery of AAV-GDNF prevents nigral neurodegeneration and promotes functional recovery in a rat model of Parkinson’s disease. Gene Ther. 2002;9:381–389.
  • Kells AP, Eberling J, Su X, et al. Regeneration of the MPTP-lesioned dopaminergic system after convection-enhanced delivery of AAV2-GDNF. J Neurosci. 2010;30:9567–9577.
  • Eslamboli A, Georgievska B, Ridley RM, et al. Continuous low-level glial cell line-derived neurotrophic factor delivery using recombinant adeno-associated viral vectors provides neuroprotection and induces behavioral recovery in a primate model of Parkinson’s disease. J Neurosci. 2005;25:769–777.
  • Sajadi A, Bensadoun JC, Schneider BL, et al. Transient striatal delivery of GDNF via encapsulated cells leads to sustained behavioral improvement in a bilateral model of Parkinson disease. Neurobiol Dis. 2006;22:119–129.
  • Shingo T, Date I, Yoshida H, et al. Neuroprotective and restorative effects of intrastriatal grafting of encapsulated GDNF-producing cells in a rat model of Parkinson’s disease. J Neurosci Res. 2002;69:946–954.
  • Decressac M, Ulusoy A, Mattsson B, et al. GDNF fails to exert neuroprotection in a rat α-synuclein model of Parkinson’s disease. Brain. 2011;134:2302–2311.
  • Kirik D, Rosenblad C, Bjorklund A, et al. Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson’s model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J Neurosci. 2000;20:4686–4700.
  • Decressac M, Kadkhodaei B, Mattsson B, et al. α-Synuclein-induced down-regulation of Nurr1 disrupts GDNF signaling in nigral dopamine neurons. Sci Transl Med. 2012;4:163ra156.
  • Gill SS, Patel NK, Hotton GR, et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat Med. 2003;9:589–595.
  • Biju KC, Santacruz RA, Chen C, et al. Bone marrow-derived microglia-based neurturin delivery protects against dopaminergic neurodegeneration in a mouse model of Parkinson’s disease. Neurosci Lett. 2013;535:24–29.
  • Gasmi M, Brandon EP, Herzog CD, et al. AAV2-mediated delivery of human neurturin to the rat nigrostriatal system: long-term efficacy and tolerability of CERE-120 for Parkinson’s disease. Neurobiol Dis. 2007;27:67–76.
  • Kordower JH, Herzog CD, Dass B, et al. Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann Neurol. 2006;60:706–715.
  • Wang W, Sun M, Li H, et al. The delivery of tyrosine hydroxylase accelerates the neurorestoration of Macaca Rhesus model of Parkinson’s disease provided by Neurturin. Neurosci Lett. 2012;524:10–15.
  • Lacomblez L, Bensimon G, Leigh PN, et al. A confirmatory dose-ranging study of riluzole in ALS. ALS/Riluzole Study Group-II. Neurology. 1996;47:S242–S250.
  • Zhang ZG, Zhang L, Jiang Q, et al. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J Clin Invest. 2000;106:829–838.
  • Shen F, Su H, Fan Y, et al. Adeno-associated viral-vector-mediated hypoxia-inducible vascular endothelial growth factor gene expression attenuates ischemic brain injury after focal cerebral ischemia in mice. Stroke. 2006;37:2601–2606.
  • Shen F, Fan Y, Su H, et al. Adeno-associated viral vector-mediated hypoxia-regulated VEGF gene transfer promotes angiogenesis following focal cerebral ischemia in mice. Gene Ther. 2008;15:30–39.
  • Wang HB, Yang L, Wu J, et al. Reduced ischemic injury after stroke in mice by angiogenic gene delivery via ultrasound-targeted microbubble destruction. J Neuropathol Exp Neurol. 2014;73:548–558.
  • Mavilio F. Gene therapies need new development models. Nature. 2012;490:7.
  • Humpel C. Identifying and validating biomarkers for Alzheimer’s disease. Trends Biotechnol. 2011 Jan;29(1):26–32.
  • Malek N, Swallow D, Grosset KA, et al. Alpha-synuclein in peripheral tissues and body fluids as a biomarker for Parkinson’s disease - a systematic review. Acta Neurol Scand. 2014;130:59–72.
  • Rothstein L, Jickling GC. Ischemic stroke biomarkers in blood. Biomark Med. 2013;7:37–47.
  • Jickling GC, Sharp FR. Blood biomarkers of ischemic stroke. Neurotherapeutics. 2011;8:349–360.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.