Dopamine Gene Therapy for Parkinson’s Disease in a Nonhuman Primate Without Associated Dyskinesia

A gene therapy approach for the treatment of Parkinson’s disease. Several high-profile patients—fighter Muhammad Ali, Attorney General Janet Reno, Pope John Paul II, and Michael J. Fox—have thrust Parkinson’s disease (PD) into the popular press in the last decade. But it was nearly 50 years ago that l-dopa was introduced as a therapy for patients with PD, and this drug, with its troublesome side effects, remains the frontline treatment for this debilitating disease that has no cure. Now, an international team of researchers describe a potential treatment for PD that uses a multigene therapy approach designed to restore continuous synthesis of the neurotransmitter dopamine in the PD brain. PD arises from the destruction of a region of the midbrain called the substantia nigra, which is part of the basal ganglia—structures in the brain that control movement and motivation. Neurons in the substantia nigra produce the neurotransmitter dopamine, a key regulator of voluntary movement, cognition, and behavior. Currently, the basis of PD therapy is to replenish the brain’s dopamine stores, which is achieved through periodic oral administration of the drug l-dopa, a blood-brain barrier–crossing dopamine precursor. Although l-dopa treatment has restored motor function in millions of PD patients, this drug does not block the progressive neurodegeneration associated with the disease and, over time, can spur troublesome side effects, such as freezing and involuntary movement. These movement-related repercussions are caused by intermittent oral delivery of l-dopa, which gives rise to peaks and valleys in brain dopamine concentrations. Thus, scientists have sought treatment approaches that deliver dopamine in a continuous manner. To this end, Jarraya et al. have designed a gene therapy protocol in which the genes that encode the key dopamine biosynthetic enzymes are introduced directly into the brain to produce a perpetual, artificial dopamine factory in neurons of the striatum, the basal ganglia nucleus that receives most of the substantia nigra–released dopamine. In normal brains, the tyrosine hydroxylase enzyme converts the amino acid tyrosine to l-dopa, which is then turned into dopamine by aromatic l-amino acid decarboxylase. Another enzyme, guanosine 5′-triphosphate cyclohydrolase 1, produces a molecule that is reduced in PD brains and is needed for efficient dopamine synthesis. Because of vector-related size constraints, genes encoding these enzymes have previously been introduced into animal models of PD in three separate viral vectors and have delivered some benefits. However, for use in the clinic, it would be preferable to use one vector that encodes all three genes. Jarraya et al. used a lentiviral vector system to create such a vector and tested it in rhesus macaque monkeys artificially induced to have PD. The results of the experiments performed by Jarraya et al. reveal that one can achieve sustained, functional concentrations of dopamine in the brains of the parkinsonian monkeys and effect an improvement in mobility and a reduction in disability within the first 6 weeks after injection of the gene-carrying vector. Most encouraging is the fact that these effects were maintained, without the troublesome involuntary movements observed in l-dopa–treated patients, for more than a year in treated animals. Although these results are promising, a number of caveats remain, including the fact that the dopamine factory introduced by gene transfer resides in striatal neurons that do not normally produce dopamine. The ongoing phase 1 and 2 clinical trial conducted by the same group represents the ultimate test of the proof-of-concept findings described in this translational study. In Parkinson’s disease, degeneration of specific neurons in the midbrain can cause severe motor deficits, including tremors and the inability to initiate movement. The standard treatment is administration of pharmacological agents that transiently increase concentrations of brain dopamine and thereby discontinuously modulate neuronal activity in the striatum, the primary target of dopaminergic neurons. The resulting intermittent dopamine alleviates parkinsonian symptoms but is also thought to cause abnormal involuntary movements, called dyskinesias. To investigate gene therapy for Parkinson’s disease, we simulated the disease in macaque monkeys by treating them with the complex I mitochondrial inhibitor 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, which induces selective degeneration of dopamine-producing neurons. In this model, we demonstrated that injection of a tricistronic lentiviral vector encoding the critical genes for dopamine synthesis (tyrosine hydroxylase, aromatic l-amino acid decarboxylase, and guanosine 5′-triphosphate cyclohydrolase 1) into the striatum safely restored extracellular concentrations of dopamine and corrected the motor deficits for 12 months without associated dyskinesias. Gene therapy–mediated dopamine replacement may be able to correct Parkinsonism in patients without the complications of dyskinesias.

[1]  Y. Agid,et al.  Effects of high-frequency stimulation on subthalamic neuronal activity in parkinsonian patients. , 2004, Archives of neurology.

[2]  A. Lang,et al.  Parkinson's disease. Second of two parts. , 1998, The New England journal of medicine.

[3]  Vesna Sossi,et al.  A double‐blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease , 2003, Annals of neurology.

[4]  Vincent Frouin,et al.  Automated Three-Dimensional Analysis of Histological and Autoradiographic Rat Brain Sections: Application to an Activation Study , 2007, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[5]  Thomas Boraud,et al.  Attenuation of levodopa-induced dyskinesia by normalizing dopamine D3 receptor function , 2003, Nature Medicine.

[6]  Kellaway Iw Scientific rationale and clinical implications of sustained-release formulations. , 1988 .

[7]  O. Andreassen,et al.  Estimation of the number of somatostatin neurons in the striatum: An in situ hybridization study using the optical fractionator method , 1996, The Journal of comparative neurology.

[8]  J. Saint-Cyr,et al.  Neuropsychological consequences of chronic bilateral stimulation of the subthalamic nucleus in Parkinson's disease. , 2000, Brain : a journal of neurology.

[9]  J. Kesslak,et al.  Transplantation of embryonic dopamine neurons for severe Parkinson's disease , 2001 .

[10]  F. Jiménez-Jiménez,et al.  Pathologic gambling in Parkinson's disease: A behavioral manifestation of pharmacologic treatment? , 2000, Movement disorders : official journal of the Movement Disorder Society.

[11]  A. Crane,et al.  Changes in local cerebral glucose utilization associated with Parkinson's syndrome induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the primate. , 1987, Life sciences.

[12]  S. Leff,et al.  Long-term restoration of striatal l-aromatic amino acid decarboxylase activity using recombinant adeno-associated viral vector gene transfer in a rodent model of Parkinson's disease , 1999, Neuroscience.

[13]  M. Hoehn,et al.  Parkinsonism , 1998, Neurology.

[14]  A. Grace Phasic versus tonic dopamine release and the modulation of dopamine system responsivity: A hypothesis for the etiology of schizophrenia , 1991, Neuroscience.

[15]  A. Björklund,et al.  Reversal of motor impairments in parkinsonian rats by continuous intrastriatal delivery of l-dopa using rAAV-mediated gene transfer , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[16]  E. Bézard,et al.  Molecular mechanisms of l-DOPA-induced dyskinesia. , 2011, International review of neurobiology.

[17]  B Bioulac,et al.  Reversal of Rigidity and Improvement in Motor Performance by Subthalamic High‐frequency Stimulation in MPTP‐treated Monkeys , 1993, The European journal of neuroscience.

[18]  N. Mazarakis,et al.  New methods to titrate EIAV-based lentiviral vectors. , 2002, Molecular therapy : the journal of the American Society of Gene Therapy.

[19]  T. Chase,et al.  Levodopa‐induced dyskinesias improved by a glutamate antagonist in parkinsonia monkeys , 1996 .

[20]  Y. Agid,et al.  Levodopa‐induced dyskinesias in Parkinson's disease: Is sensitization reversible? , 2000 .

[21]  A. Kingsman,et al.  Stable gene transfer to the nervous system using a non-primate lentiviral vector , 1999, Gene Therapy.

[22]  J. Bloch,et al.  Lentiviral Gene Transfer to the Nonhuman Primate Brain , 1999, Experimental Neurology.

[23]  M. Matsumura,et al.  Behavioral recovery in a primate model of Parkinson's disease by triple transduction of striatal cells with adeno-associated viral vectors expressing dopamine-synthesizing enzymes. , 2002, Human gene therapy.

[24]  N. Quinn,et al.  L-dopa dose and the duration and severity of dyskinesia in primed MPTP-treated primates , 2007, Journal of Neural Transmission.

[25]  T. Creighton Methods in Enzymology , 1968, The Yale Journal of Biology and Medicine.

[26]  G. Fénelon,et al.  Hallucinations in Parkinson's disease: prevalence, phenomenology and risk factors. , 2000, Brain : a journal of neurology.

[27]  H. Mizukami,et al.  Triple transduction with adeno-associated virus vectors expressing tyrosine hydroxylase, aromatic-L-amino-acid decarboxylase, and GTP cyclohydrolase I for gene therapy of Parkinson's disease. , 2000, Human gene therapy.

[28]  E. Bézard,et al.  Comparison of eight clinical rating scales used for the assessment of MPTP-induced parkinsonism in the Macaque monkey , 2000, Journal of Neuroscience Methods.

[29]  U Sabatini,et al.  Supplementary and primary sensory motor area activity in Parkinson's disease. Regional cerebral blood flow changes during finger movements and effects of apomorphine. , 1992, Archives of neurology.

[30]  A. Benabid,et al.  Electrical stimulation of the subthalamic nucleus in advanced Parkinson's disease. , 1998, The New England journal of medicine.

[31]  Haruhiko Kishima,et al.  Functional Recovery in a Primate Model of Parkinson's Disease following Motor Cortex Stimulation , 2004, Neuron.

[32]  C. Tanner,et al.  Levodopa and the progression of Parkinson's disease. , 2004, The New England journal of medicine.

[33]  L. Sokoloff,et al.  The [14C]deoxyglucose method: four years later. , 1979, Acta neurologica Scandinavica. Supplementum.

[34]  Werner Poewe,et al.  Treatment interventions for Parkinson's disease: an evidence based assessment , 2002, The Lancet.

[35]  B Bioulac,et al.  Dopamine agonist-induced dyskinesias are correlated to both firing pattern and frequency alterations of pallidal neurones in the MPTP-treated monkey. , 2001, Brain : a journal of neurology.

[36]  R. Lloyd Four years later , 1999 .

[37]  P. Pollak,et al.  Transient acute depression induced by high-frequency deep-brain stimulation. , 1999, The New England journal of medicine.

[38]  R. Mark Wightman,et al.  Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine transporter , 1996, Nature.

[39]  L. Metman,et al.  Effects of supra-threshold levodopa doses on dyskinesias in advanced Parkinson's disease , 1997, Neurology.

[40]  Guo-Rong Zhang,et al.  Coexpression of tyrosine hydroxylase, GTP cyclohydrolase I, aromatic amino acid decarboxylase, and vesicular monoamine transporter 2 from a helper virus-free herpes simplex virus type 1 vector supports high-level, long-term biochemical and behavioral correction of a rat model of Parkinson's disease. , 2004, Human gene therapy.

[41]  Fabrizio Stocchi,et al.  Continuous dopamine-receptor treatment of Parkinson's disease: scientific rationale and clinical implications , 2006, The Lancet Neurology.

[42]  M. Cenci,et al.  Post‐ versus presynaptic plasticity in L‐DOPA‐induced dyskinesia , 2006, Journal of neurochemistry.

[43]  J. Dostrovsky,et al.  Microstimulation-induced inhibition of neuronal firing in human globus pallidus. , 2000, Journal of neurophysiology.

[44]  A. Lang,et al.  Parkinson's disease. First of two parts. , 1998, The New England journal of medicine.

[45]  Y Agid,et al.  Transient acute depression induced by high-frequency deep-brain stimulation. , 1999, The New England journal of medicine.

[46]  J. Nutt,et al.  Effect of long-term therapy on the pharmacodynamics of levodopa. Relation to on-off phenomenon. , 1992, Archives of neurology.

[47]  C. Legéndy,et al.  Bursts and recurrences of bursts in the spike trains of spontaneously active striate cortex neurons. , 1985, Journal of neurophysiology.

[48]  M. Grégoire,et al.  Anatomic and Biochemical Correlates of the Dopamine Transporter Ligand 11C-PE2I in Normal and Parkinsonian Primates: Comparison with 6-[18F]Fluoro-L-Dopa , 2001, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[49]  Anatol C. Kreitzer,et al.  Striatal Plasticity and Basal Ganglia Circuit Function , 2008, Neuron.

[50]  A. Kingsman,et al.  Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery. , 2001, Human molecular genetics.

[51]  Murtaza Z Mogri,et al.  Optical Deconstruction of Parkinsonian Neural Circuitry , 2009, Science.

[52]  A. Kingsman,et al.  A transient three-plasmid expression system for the production of high titer retroviral vectors. , 1995, Nucleic acids research.

[53]  P. Pivirotto,et al.  Long-term clinical improvement in MPTP-lesioned primates after gene therapy with AAV-hAADC. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[54]  A. Björklund,et al.  Reversal of dyskinesias in an animal model of Parkinson's disease by continuous L-DOPA delivery using rAAV vectors. , 2005, Brain : a journal of neurology.

[55]  R. Cools Dopaminergic modulation of cognitive function-implications for l-DOPA treatment in Parkinson's disease , 2006, Neuroscience & Biobehavioral Reviews.

[56]  A. Kingsman,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. , 2002, The Journal of Neuroscience.

[57]  J. Bloch,et al.  Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease. , 2000, Science.

[58]  Anders Björklund,et al.  Cell transplantation in Parkinson's disease: how can we make it work? , 2005, Trends in Neurosciences.

[59]  P. Sanders Evidence based assessment , 1996, British Dental Journal.