Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Parkinson’s Disease: Gene Therapies

Philippe G. Coune, Bernard L. Schneider, and Patrick Aebischer


Neurodegenerative Studies Laboratory, Brain Mind Institute, Ecole Polytechnique Fédérale
de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
Correspondence: patrick.aebischer@epfl.ch

With the recent development of effective gene delivery systems, gene therapy for the central
nervous system is finding novel applications. Here, we review existing viral vectors and dis-
cuss gene therapy strategies that have been proposed for Parkinson’s disease. To date, most of
the clinical trials were based on viral vectors to deliver therapeutic transgenes to neurons with-
in the basal ganglia. Initial trials used genes to relieve the major motor symptoms caused by
nigrostriatal degeneration. Although these new genetic approaches still need to prove more
effective than existing symptomatic treatments, there is a need for disease-modifying strat-
egies. The investigation of the genetic factors implicated in Parkinson’s disease is providing
precious insights in disease pathology that, combined with innovative gene delivery systems,
will hopefully offer novel opportunities for gene therapy interventions to slow down, or even
halt disease progression.
www.perspectivesinmedicine.org

arkinson’s disease (PD) presents several fea- to the basal ganglia nuclei are currently investi-
P tures that make it an ideal target for gene
therapy. PD is a progressive, long-lasting neu-
gated to possibly halt disease progression.

rodegenerative disease that is not adequately


PRINCIPLE AND TOOLS
treated with pharmacological approaches. Fur-
thermore, the cause of the major motor symp- Gene therapy aims at treating disease by geneti-
toms is well defined, and associated with dys- cally modifying populations of cells that are
functions in brain nuclei that can be effectively either directly functionally impaired or capable
targeted with viral vector technologies for gene of relieving the disease symptoms. These gene-
transfer to brain cells. Therefore, part of the tic modifications can either increase or reduce
gene therapy strategies aim at increasing the effi- the expression of specific genes or gene sets, or
cacy and at reducing the side effects of the cur- even restore the normal function of the product
rent pharmacological and surgical treatments. of these genes. In human therapeutic applica-
Besides the possibility to correct motor sym- tions, these modifications can only be made on
ptoms, our understanding of the genetic causes somatic cells and must spare the germline for
of PD has made considerable progress during ethical reasons. A distinction is made between
the last decade. Consequently, several strategies in vivo gene therapy, i.e., the direct genetic mod-
based on the delivery of neuroprotective genes ification of cells inside the body, and ex vivo gene

Editor: Serge Przedborski


Additional Perspectives on Parkinson’s Disease available at www.perspectivesinmedicine.org
Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a009431
Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431

1
P. Coune et al.

therapy, which is based on the genetic modifica- proteins, and cis-acting factors controlling the
tion of cells maintained in culture prior to im- packaging, reverse transcription, nuclear local-
plantation in the patient. ization and integration of the viral genome in
Various methods have been developed for the host cell genome. Lentivectors have been
gene delivery to the target cells, which include vi- developed from primate lentiviruses, such as
ral vectors, and nonviral systems. Nonviral meth- the wild-type human immunodeficiency virus
ods, which are marginally used for gene transfer type 1 (HIV-1) and nonprimate lentiviruses,
to the central nervous system (CNS), comprise such as the equine infectious anemia virus
chemical and physical methods, such as gene (EIAV), by progressively removing most of the
gun or electroporation. In vivo gene transfer us- viral genes from the vector genome to limit
ing viral vectors is today the most commonly the risk of producing replication-competent vi-
used approach in the CNS, with 20 trials listed ral particles (Naldini et al. 1996b; Zufferey et al.
in 2010 (Lim et al. 2010). This approach takes 1997, 1998; Dull et al. 1998; Olsen 1998). De-
advantage of the viruses’ ability to deliver their pending on the production system employed,
genetic material to target cells, including nondi- the viral genes gag, pol, tat, and rev, which are
viding cells, and to induce long-term transgene necessary to the production of infectious viral
expression. As most of the cells in the CNS, includ- particles, are provided in trans using separate
ing neurons, are postmitotic, the ability of viral plasmids cotransfected in the packaging cells.
vectors to transduce nondividing cells is of crucial The maximal packaging capacity of lentivec-
importance in the context of PD gene therapy. tors is 18 kb, but the most efficient packaging
Viral vectors areengineered fromwild-typevi- is obtained with genomes in the range of 6 to
ruses by removing the genes essential to their rep- 9 kb (Kumar et al. 2001). To target neurons
lication from their genome. The removed genetic and obtain highly concentrated vector suspen-
www.perspectivesinmedicine.org

information is provided in trans for vector pro- sions, lentivectors are typically pseudotyped
duction, and is not incorporated inside the par- by replacing the wild-type envelope with the en-
ticles. The vectors are therefore able to infect cells velope of the vesicular stomatitis virus G (VSV-
and transfer their genetic material into the nu- G). The resulting pseudotyped vectors have
cleus, but they are unable to replicate themselves a broad cell tropism including neuronal and
in the host cells. This aspect is critical for vector glial cells (Naldini et al. 1996a). Lentivectors
biosafety, as it eliminates virus pathogenicity have the ability to integrate into the host cell ge-
and prevents uncontrolled spreading of transgene nome and lead to stable transgene expression.
delivery caused by vector replication in the host This feature is particularly useful for ex vivo
organism. Although viral gene delivery systems gene therapy applications: cell lines or stem
lead to irreversible genetic modifications within cells can be stably transduced using lentivectors
the patient tissues, currently available vectors al- and transgene expression carefully character-
low for efficient and stable transgene expression ized prior to implantation. Because integration
in a broad range of cellular types across the brain. occurs at random sites, there is a risk for inser-
Several types of vectors have been developed, dif- tional mutagenesis that has to be considered
fering by their packaging capacity, tropism, and for in vivo gene therapy. However, a study
immunogenicity. Herewewill focus onvectors de- using lentivectors in a mouse strain highly sus-
rived from adeno-associated virus (AAV) and len- ceptible to tumor formation did not reveal any
tivirus, as they are the only ones that have reached increase in tumor occurrence (Montini et al.
the clinic for CNS gene therapy trials. 2006), and an on-going clinical trial using
lentivectors in PD did not report any evidence
of tumorigenicity yet (Lim et al. 2010). To ad-
Lentivectors
dress this potential problem, nonintegrating
Lentiviruses are enveloped viruses from the lentiviral vectors have been developed to im-
Retroviridae family. Their genome consists of prove the vector safety profile (Rahim et al.
single-stranded RNA that encodes structural 2009).

2 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

AAV-Based Vectors and display a good neuronal tropism (Paterna


et al. 2004). They enter the host cell though re-
Vectors derived from adeno-associated viruses ceptor-mediated endocytosis, and are translo-
(AAV) are the most frequent type of vectors cated to the nucleus where their genome is con-
used in clinical trials for CNS diseases. AAVs verted into double-stranded DNA. Although
are small nonenveloped viruses of the parvovir- wild-type AAVs preferentially integrate to the
idae family. The 4.7 kb AAV genome is com- human genome at a specific locus on chromo-
posed of single-stranded DNA encoding four some 19, termed AAVS1 (Samulski et al. 1991;
proteins essential for replication and packaging Huser et al. 2010), recombinant AAV vectors
(rep gene), and three capsid proteins (cap gene), form stable episomal concatemers in the absence
surrounded by two inverted terminal repeats of the Rep protein. In postmitotic cells, episomal
(ITR). Even in their wild-type form, AAVs de- AAV genomes provide long-term (.1 yr) trans-
pend on the coinfection with a helper virus, gene expression. The low rate of integration of
such as adenovirus or herpes virus, for efficient recombinant AAV vectors is considered an asset
replication inside the host cells. As AAVs are in their safety profile, as it limits the occurrence
considered nonpathogenic in humans, and their of insertional mutagenesis.
capsid proteins induce only mild immune reac- There are currently more than 100 AAV se-
tions (Büning et al. 2008), they are prime can- rotypes described from different animal spe-
didates for gene therapy. However, 90% of the cies (Kwon and Schaffer 2008). These serotypes
population has been exposed to AAV serotype mainly differ by the protein composition of the
2, and therefore has preexisting antibodies that capsid. As AAV tropism is mainly determined by
will neutralize AAV2-based vectors and reduce the properties of the capsid, it is possible to cre-
their transduction efficacy (Moskalenko et al. ate pseudotyped vectors with various tropisms
www.perspectivesinmedicine.org

2000). Genetic modifications of the capsid pro- by packaging the genome of a particular sero-
teins can disrupt the immunogenic epitopes and type (most often AAV2) into a capsid derived
possibly circumvent this limitation (Maersch from another serotype.
et al. 2010). In addition, the CNS benefits from Overall, AAV vectors represent ideal vectors
a relative immune privilege, which reduces the to deliver genes in the CNS. They present a very
risk of neutralizing activity. good safety profile, and provide efficient trans-
Viral vectors have been derived from wild- duction and durable expression of neurons. The
type AAVs by the deletion of all the viral sequen- main limitation of AAV is the limited packaging
ces except the ITRs. Consequently, AAV vectors capacity (4.7 kb) that precludes the integration
cannot replicate and do not express any viral of large genes, or multiple expression cassettes.
proteins, which further reduces their immuno-
genicity. During the production of AAV vectors,
SYMPTOMATIC THERAPIES
rep and cap are typically provided in trans using a
helper plasmid system cotransfected with the Current pharmacological and surgical therapies
transgene plasmid in presence of an adenovirus for PD all aim at compensating for the basal gan-
(Samulski et al. 1989). More recently, “helper vi- glia dysfunction caused by the degeneration of
rus-free” systems have been developed, in which the dopaminergic neurons from the substantia
the adenoviral helper activity is also provided via nigra pars compacta (SNpc). L-dopa therapy in-
the recombinant helper plasmid (Grimm et al. creases dopamine production in the remaining
2003). After transfection, recombinant AAV par- nigral neurons and thereby compensates for
ticles are harvested by lysis of the packaging cells, the lack of neurotransmitter in the striatum.
purified, and finally concentrated in suspen- On the other hand, deep brain electrical stimu-
sions that typically contain 1012 – 1014 particles lation (DBS) modulates the overactivity of the
per ml. subthalamic nucleus consecutive to the loss of
AAV vector particles can efficiently infect a dopamine signaling in the striatum. Although
broad range of cells, including postmitotic cells, both approaches provide symptomatic relief to

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 3


P. Coune et al.

PD patients, they also present a number of draw- the apparition of dyskinesia and limit the side ef-
backs. Gene therapy approaches have been inves- fects caused by elevated dopamine levels outside
tigated for their ability to similarly correct motor the basal ganglia (Fig. 1).
symptoms and possibly reduce the occurrence
of adverse effects associated with the existing
Full Ectopic Dopamine Synthesis
symptomatic treatments.
A possible approach is to deliver the genes TH,
AADC, and GCH1 to medium spiny neurons
Enzyme Replacement Strategies
(MSN) in the striatum, to induce ectopic dopa-
Similar to L-dopa-therapy, enzyme replacement mine synthesis from tyrosine. This strategy has
therapies aim at compensating for the decrease been shown to successfully increase striatal dop-
of dopamine release resulting from the degen- amine and correct motor deficits in 6-OHDA le-
eration of the nigrostriatal dopaminergic neu- sioned rats (Shen et al. 2000; Sun et al. 2003) and
rons. These approaches are based on the transfer MPTP-treated cynomolgus macaque monkeys
of genes encoding the enzymes required for do- (Muramatsu et al. 2002). However, elevated con-
pamine synthesis into the striatal GABAergic centrations of dopamine can negatively regulate
neurons. Endowed with these enzymes, the TH activity via a feedback loop that may limit the
GABAergic neurons are able to ectopically syn- capacity for ectopic dopamine synthesis. This
thetize dopamine that will be released in the caveat can be circumvented by the use of a con-
striatum. stitutively active truncated version of TH (Mof-
Dopamine is synthesized from tyrosine, fat et al. 1997).
which is provided either by the nutrition or A tri-cistronic EIAV-based lentivector cod-
through the conversion of phenylalanine into ty- ing for AADC and GCH1 and a truncated form
www.perspectivesinmedicine.org

rosine by the enzyme phenylalanine hydroxylase. of TH, has been developed by Oxford BioMedica
Tyrosine is next hydroxylated into L-dopa by ty- under the name of ProSavin and successfully
rosine hydroxylase (TH) and finally, L-dopa is tested in 6-OHDA-lesioned rats (Azzouz et al.
converted into dopamine by the aromatic amino 2002). A study in MPTP-treated macaque mon-
acid decarboxylase (AADC). TH activity is the keys (Jarraya et al. 2009) showed that ProSavin
limiting factor in dopamine biosynthesis, and re- treatment does not only reduce motor impair-
quires a cofactor, tetrahydrobiopterine (BH4), ment, but also leads to a drastic reduction of
the synthesis of which requires the enzyme dyskinesia when compared to conventional
GTP-cyclohydrolase-1 (GCH-1) (Kettler et al. L-dopa-therapy. The safety, efficacy, and dose
1974). Synthetized dopamine is transferred and evaluation of ProSavin are currently under eval-
concentrated in storage vesicles by the vesicular uation in a phase I/II clinical trial.
monoamine transporter VMAT2.
Although L-dopa therapy has been the
Ectopic L-dopa Conversion
mainstay of PD therapy since 1969 (Cotzias et al.
1969), its efficacy is limited by major side effects. Genetic enzyme replacement therapy can also
Because dopaminergic drugs diffuse poorly into be used to increase the efficacy of pharmacolog-
the CNS, they need to be systemically adminis- ical L-dopa therapy. The degeneration of the
tered at high dose with an increased risk of pe- nigral dopaminergic neurons leads to a decrease
ripheral adverse effects. At advanced stages of in striatal AADC activity, which is essential for
the disease, discontinuous L-dopa therapy is as- L-dopa conversion into dopamine (Ichinose
sociated with the apparition of debilitating dys- et al. 1994). Therefore, the efficacy of the L-
kinesia presumably linked to fluctuations in the dopa treatment can be eventually limited by
brain concentration of L-dopa. Gene delivery of the loss of AADC activity. Advanced PD patients
the enzymes needed for dopamine synthesis could benefit from gene transfer to restore stria-
could provide a continuous ectopic production tal AADC levels, thereby lowering the effective
of dopamine in the striatum, which may reduce dose of L-dopa and limiting the side effects.

4 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

Tyrosine
Tyrosine L-dopa HO
HO HO NH2 = Provided by nutrition
NH2 NH2
COOH
COOH HO COOH

TH + AADC L-dopa
GCH1 HO
NH2
Dopamine = Provided by medication
L-dopa HO HO COOH
NH2
HO
NH2
HO
HO COOH
Ectopic dopa
AADC conversion Dopamine
Dopamine Tyrosine HO
NH2
HO
HO NH2
NH2 HO
COOH
HO L-dopa
TH + AADC
Complete ectopic GCH1 HO
NH2
dopamine synthesis
L-dopa HO COOH
HO
NH2
AADC compartment
HO COOH

Medium Ectopic dopa


spiny neuron synthesis
www.perspectivesinmedicine.org

Figure 1. The different enzyme replacement therapy approaches in PD. Enzyme replacement therapies aim at
compensating the decrease of striatal dopamine release caused by the degeneration of the nigral dopaminergic
neurons, by inducing ectopic dopamine synthesis in the striatum.

AAV vectors have been designed to deliver the AADC striatal expression allows for a control of
AADC coding sequence to the striatal MSNs, the gene therapy effects via the modulation of
which are then able to convert the adminis- the L-dopa dosage. As systems for controlling
tered L-dopa into dopamine. Striatal injection the level of transgene expression are not current-
of AADC-coding AAV vectors was shown to ly available for human applications, such a com-
effectively and stably restore the response to binedapproachofferssomeadvantagesoverother
L-dopa in MPTP-treated rhesus macaque mon- nonregulated gene therapy approaches for PD.
keys (Bankiewicz et al. 2000, 2006). A phase I Strategies aiming at chronic ectopic dopa-
clinical trial (Eberling et al. 2008; Christine mine production in the striatum may cause det-
et al. 2009) in bilaterally infused patients did rimental effects over time. Indeed, these gene
not report any significant adverse effects related therapy approaches target GABAergic striatal
to the viral vector itself, even though the stereo- neurons that are unable to store dopamine into
taxic surgery led to hemorrhages in several pa- vesicles for synaptic release. Ectopic dopamine
tients. PET imaging reported an increase of synthesis may therefore lead to uncontrolled ex-
striatal AADC activity at 1 and 6 months postin- tracellular or cytosolic levels of dopamine, often
jection, which was dependent on the vector dose associated with chronic oxidative stress. Increase
administered and confirmed stable transgene in both cytosolic (Chen et al. 2008) and extracel-
expression. Modest clinical improvements were lular (Cyr et al. 2003) levels of dopamine have
observed in both on- and off-states, in addition been shown to lead to the degeneration of the
to a decrease of the effective L-dopa dose. How- striatal neurons. Therefore, therapeutic strat-
ever, because of the open design of the trial, egies should be cautiously designed to avoid ex-
placebo effect cannot be excluded. Importantly, cessive striatal dopamine concentration.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 5


P. Coune et al.

Ectopic L-dopa Synthesis crease in the inhibitory control exerted by the


external segment of the globus pallidus (GPe)
An alternative gene therapy approach aims at
on the subthalamic nucleus (STN). The subse-
ectopically producing L-dopa in the striatum
quent lack of inhibition of the STN affects the
by transfer of the TH and GCH1 genes into
output of the basal ganglia circuitry, thereby
MSNs. Endogenous AADC activity can convert
causing impairments in motor functions. DBS
the L-dopa produced in the striatum into dopa-
is based on high frequency electrical stimula-
mine.
tions applied to the STN to interfere with its ab-
This strategy has been shown to improve
normally high activity. By doing so, DBS can
motor deficits in rats presenting partial (Kirik
significantly improve the main motor parkinso-
et al. 2002) and total 6-OHDA-induced nigros-
nian symptoms.
triatal denervation (Björklund et al. 2010), and
In 2002, Luo et al. described a gene therapy
to reduce dyskinesia caused by intermittent ad-
approach aiming at modulating STN activity
ministrations of L-dopa (Carlsson et al. 2005).
(Luo et al. 2002) by delivering the genes glutamic
Interestingly, 11C-raclopride positron emission
acid decarboxylase (GAD), the rate-limiting en-
tomography (PET) analyses indicates that func-
zyme for the synthesis of GABA. Using an AAV
tional recovery is correlated with the recon-
vector, the approach targets the glutamatergic
stitution of a functional pool of endogenous do-
neurons of the STN. The neurons transduced
pamine in the striatum (Leriche et al. 2009).
with GAD synthesize and release GABA in an
This finding is surprising considering that there
activity-dependent manner. The modified neu-
is no evidence that dopamine storage and re-
rotransmitter pattern in STN projections leads
lease mechanisms are restored. Studies in non-
to an inhibitory bias in the substantia nigra
human primates are currently ongoing to fur-
pars reticulata (SNr) and regulates the firing
www.perspectivesinmedicine.org

ther assess the safety and efficacy of this gene


rates of basal ganglia nuclei in 6-OHDA-les-
therapy strategy.
ioned rats (Lee et al. 2005). These effects on the
The loss of AADC activity caused by the
biochemical and electrophysiological properties
progression of nigrostriatal neurodegeneration
of the STN-to-SNr projections improve the
may limit the long-term efficacy of this approach.
6-OHDA-induced motor deficits in rats as a
However, striatal serotoninergic terminalsprojec-
function of GAD expression levels (Luo et al.
ting from the raphe nucleus do not degenerate to
2002; Lee et al. 2005). These findings were later
the same extent as dopaminergic terminals in PD
confirmed in MPTP-treated rhesus monkeys.
(Kish et al. 2008). As they express both AADC and 18
F-fluorodeoxyglucose (FDG) PET investiga-
VMAT2, they are likely to provide a disease-resist-
tion showed that the improvement in clinical
ant source of AADC activity for L-dopa conver-
ratings is correlated with an increase in glucose
sion into dopamine. In addition, preclinical stud-
metabolism in the ipsilateral motor cortex (Em-
ies have shown that even in case of serotoninergic
borg et al. 2007). However, although GAD ex-
denervation, there is enough remaining striatal
pression in the STN of 6-OHDA lesioned rats
AADC activity for dopamine production. Al-
had a neuroprotective effect (Luo et al. 2002),
though the exact nature of that third AADC com-
no such effect was observed in the MPTP-treated
partment is currently unknown, it may include
monkeys. Importantly, these preclinical studies
striatal neurons and nonneuronal cells, such as as-
did not report any adverse effects in animal
trocytes (Björklund et al. 2010).
models.
Clinical studies were initiated in PD patients.
Glutamic Acid Decarboxylase Expression
In mammals, there are two isoforms of GAD,
in the Subthalamic Nucleus
GAD65, and GAD67, which are both expressed
In PD, the loss of dopamine signaling in the in most GABAergic neurons in the brain (LeWitt
striatum affects the activity of several deep brain et al. 2011). As these two isoforms displayed var-
nuclei. In particular, striatal dopamine deple- iable relative efficacy in rats and primate models
tion leads, via the “indirect” pathway, to a de- of PD, it was chosen to conduct human trials

6 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

using a combination of two AAV2 vectors, each DISEASE-MODIFYING APPROACHES


coding for one of these isoforms.
The main therapies available for the treatment
An open-label nonrandomized stage I study
of PD aim at restoring dopamine levels or cor-
investigated the safety and tolerability of unilat-
recting the functional perturbations of the basal
eral subthalamic injection of AAV2-GAD65/67
ganglia caused by dopamine loss. However, these
in 12 patients (Kaplitt et al. 2007). Using three
symptomatic therapies do not slow down dis-
vector doses, no adverse effects related to the
ease progression, and their efficacy therefore
procedure and no change in the patients’ im-
declines over time. During the last decade, the
munity against AAVor GAD were reported after
discovery of genes linked to familial forms of
12 months. As reflected by UPDRS ratings of
PD has dramatically improved our understand-
the contralateral limb, mild clinical improve-
ing of the possible molecular causes of neurode-
ments were found in both on- and off-states as
generation.
soon as 3 months postsurgery, persisting for 12
Based on these findings, novel approaches
months. FDG-PET analysis revealed that GAD
have been proposed to possibly protect neuronal
expression in the STN corrects the thalamic
functions and halt disease progression. In this
hypermetabolism associated with PD, and in-
context, gene therapy offers an attractive alterna-
creases metabolic activity in the ipsilateral mo-
tive to deliver genetic information in the CNS
tor cortical regions (Feigin et al. 2007).
and chronically provide the needed neuroprotec-
Although the contribution of the placebo
tive effects. Here we will discuss the use of neuro-
effect remained unclear in the phase I clinical
trophic factors to support the function and sur-
trial, this first application to humans confirmed
vival of nigral dopaminergic neurons, as well as
that the procedure was safe. A phase II random-
therapeutic approaches to correct the genetic
ized, multicentric trial was therefore conducted
www.perspectivesinmedicine.org

defects associated with familial forms of PD.


to assess treatment efficacy. The trial involved 16
patients bilaterally infused with AAV2-GAD65/
67 and 21 sham-operated patients, monitored Neurotrophic Factors: GDNF
for over 6 months. At the end of the trial, no ma- Neurotrophic factors are secreted proteins play-
jor adverse effects related to the operation were ing essential roles in the differentiation, growth,
reported, and patients displayed a modest but and survival of neuronal cells. In particular,
significant improvement of the UPDRS motor the glial cell line-derived neurotrophic factor
score (LeWitt et al. 2011). (GDNF), a member of the transforming growth
Overall, these trials have confirmed the safety factor (TGF)-b superfamily, has emerged as a
of the GAD gene transfer approach in the STN, powerful factor to protect the dopaminergic
and reported beneficial clinical effects. However, neuronal function and has therefore been in-
the improvements obtained have been so far tensely investigated as a promising target for
modest—therefore, this approach should be PD therapy. GDNF was identified in 1993 for
compared with DBS in terms of safety and effi- its potent trophic activity on dopaminergic neu-
cacy before any large-scale application could be rons in vitro (Lin et al. 1993). Since then, GDNF
envisioned. has been shown to promote axonal sprouting in
In conclusion, the exploration of sympto- vivo (Beck et al. 1995) and to be essential for the
matic gene therapies for PD has provided en- survival of dopaminergic neurons in the adult
couraging, preclinical and clinical results and brain (Pascual et al. 2008).
has showed that current viral vectors technology
allow for safe gene delivery within the basal
Direct GDNF Injections
ganglia nuclei. The availability of safe and effi-
cient vectors is crucial to the ultimate goal of As the recombinant protein GDNF does not
PD gene therapy: the development of therapies cross the human blood-brain barrier (Deierborg
that would interfere with the processes underly- et al. 2008), it is crucial to find alternatives to
ing the disease. systemic administration for PD treatment. The

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 7


P. Coune et al.

local delivery of recombinant GDNF protein adenovirus, lentivirus and AAV-based vectors to
into CNS has been investigated. Direct injec- express GDNF in the striatum or the SNpc (for
tions, or infusion of GDNF using minipumps review, see Deierborg et al. 2008). The delivery
into the striatum, midbrain, or ventricles pro- of the GDNF gene in both brain areas leads to
vided encouraging results in toxin-based rodent a reduction of dopaminergic cells loss and to be-
and primate models of PD (Kearns and Gash havioral recovery. The beneficial effect of viral
1995; Sauer et al. 1995; Grondin et al. 2002, GDNF expression on nigrostriatal degeneration
2003; Ai et al. 2003). Notably, it has been found and motor deficits was confirmed in MPTP-
that GDNF can induce the sprouting of dopami- treated rhesus monkeys (Kordower et al. 2000).
nergic axons near the site of GDNF delivery However, the toxin-based models used to ini-
(Rosenblad et al. 1999; Kirik et al. 2000). Based tially validate GDNF neuroprotective effects do
on the preclinical results, a randomized double- not provide a definite answer. The 6-OHDA
blind clinical study was initiated using intra- and MPTP toxins induce an acute degenera-
cerebroventricular GDNF infusion (Nutt et al. tion of the nigrostriatal system, which is mainly
2003). The results were disappointing, with no caused by the production of reactive oxygen
clinical improvement and adverse effects, in- species, and fails to replicate the complex patho-
cluding anorexia. The postmortem analysis of genic mechanisms involved in genetic and spo-
one of the participant confirmed the absence radic forms of PD.
of neuroprotection and revealed the absence of GDNF gene delivery has also been success-
GDNF diffusion into the brain parenchyma fully evaluated in aged monkeys, a more physio-
(Kordoweret al. 1999). Therefore, it appeared es- logical model that displays naturally occurring
sential to change the mode of GDNF delivery. mild reduction of striatal dopamine and motor
Indeed, a subsequent pilot study using striatal deficits (Kordower et al. 2000; Palfi et al. 2002;
www.perspectivesinmedicine.org

infusion with a minipump system reported Johnston et al. 2009). Importantly, the effect
positive clinical results (Gill et al. 2003). How- of GDNF has also been investigated in a rat
ever, a double-blind phase II clinical trial run genetic model of PD based on the nigral overex-
by Amgen, failed to confirm these results, and pression of wild-type and mutated forms of
the company decided to abandon further clini- a-synuclein using lentiviral (Lo Bianco et al.
cal investigation of direction GDNF infusion in 2004a) and AAV vectors (Decressac et al. 2011).
the CNS. Nevertheless, these trials provided a Surprisingly, GDNF fails to provide any neu-
strong rationale for gene therapy as a potential roprotective effect or induce striatal axonal
alternative to the intraparenchymal delivery of sprouting in nigral neurons overexpressing a-
recombinant neurotrophic factors. synuclein for reasons that remain to be eluci-
dated. Although the effect of GDNF gene ther-
apy is still investigated preclinically, two clinical
In Vivo GDNF Gene Delivery
trials have been conducted with an AAV2 vector
In contrast to the delivery of the recombinant expressing the neurotrophic factor neurturin,
protein, gene transfer allows for a constant a close homolog of GDNF. Preclinical studies
and local administration of GDNF, which limits showed that intrastriatal injections of the AAV2-
the risks of side effects associated with broad neurturin vector protect nigral neurons from
distribution of this factor, such as loss of weight 6-OHDA-induced degeneration and preserves
and allodynia (Hoane et al. 1999). As genetically animal motor behavior (Gasmi et al. 2007a,b).
modified cells continuously synthesize GDNF, A study in MPTP-treated monkeys reported mo-
this approach would avoid protein stability is- tor dysfunction prevention and confirmed neu-
sues and the potential need for repetitive inter- rturin neuroprotective effects (Kordower et al.
ventions to inject recombinant GDNF or reload 2006). Based on these encouraging results, the
implanted minipumps. bilateral intra-putaminal injection of the AAV2-
Several in-vivo studies have been conducted neuturin (CERE-120) vector was evaluated for
in toxin-based mice and rat models of PD using safety and efficacy in an open label phase I

8 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

clinical trial initiated by Ceregene (Marks et al. provide new clues for genetic strategies that
2008). The monitoring of 12 treated PD patients aim at slowing down the degeneration of vul-
did not reveal any serious adverse effects, and nerable populations of neurons, in both familial
showed a marked clinical improvement in off- and sporadic forms of PD.
medication symptoms. However, the double-
blind randomized phase II clinical study that fol-
a-Synuclein
lowed with 58 PD patients (Marks et al. 2010) did
not reveal any significant beneficial effects of The small protein a-synuclein is considered as a
AAV2-neurturin against the control group of pa- major actor in both sporadic and familial cases
tients who received sham surgery. As a possible of PD. Missense mutations in the a-synuclein
clue for the lack of effect, postmortem analysis coding sequence were initially found in rare fam-
of PD brain tissue from the AAV2-neurturin ilies with autosomal dominant PD. Interestingly,
study showed neurturin expression only in the similar cases of PD were associated with poly-
striatum, in contrast to a previous successful morphisms in the a-synuclein promoter (Mara-
study in monkeys in which the retrograde trans- ganore et al. 2006), and multiplications of the
duction of nigral dopaminergic cell bodies was locus carrying the a-synuclein gene (Singleton
observed (Bartus et al. 2011). et al. 2003; Nishioka et al. 2006). This last finding
Using viral vectors for the delivery of neuro- clearly links certain forms of PD with the ex-
trophic factors, there is a distinct risk that viral pression level of the protein. One can therefore
particles could diffuse away from the site of assume that strategies to down-regulate a-synu-
injection and elicit ectopic GDNF expression, clein expression may impact on the disease
which unpredictable consequences. Because of process. Several viral vector-based gene delivery
these safety concerns, alternative systems for ex systems have been explored to interfere with a-
www.perspectivesinmedicine.org

vivo gene transfer have been explored. The im- synuclein expression. They mainly rely on RNA
plantation of genetically engineered stem cells interference (RNAi) to selectively destabilize
(Akerud et al. 2001; Park 2001) or astrocytes the a-synuclein mRNA and/or block protein
(Cunningham and Su 2002) has been proposed. translation, via the transgenic expression of
To further improve the safety of the approach short hairpin RNAs (shRNA), or micro RNAs
and protect the grafted cells from potential (miRNA)directedagainst the a-synuclein mRNA
host immune reactions, it is possible to implant sequence. An alternative approach based on the
cells genetically engineered for GDNF expres- AAV-mediated delivery of an anti-a-synuclein
sion within a permeable polymer capsule (Tseng ribozyme has also been investigated both in vitro
et al. 1997; Kishima et al. 2004; Sajadi et al. 2006). and in vivo (Hayashita-Kinoh et al. 2006).
However, the efficacy of these approaches has RNAi has been shown to successfully reduce
not been investigated in PD patients yet. the level of both endogenous or overexpressed a-
In conclusion, the potential regenerative synuclein, either in vitro (Fountaine and Wade-
and neuroprotective effects of neurotrophic fac- Martins 2007; Junn et al. 2009) or in vivo (Sapru
tors justify further investigation in PD. However, et al. 2006; McCormack et al. 2010). In vitro
it is essential to evaluate the efficacy of this ap- silencing of A53T a-synuclein in NS20Y cells
proach on the genetic factors of PD. was found to decrease proteasome impairment
caused by a-synuclein and increase the cell
resistance to oxidative stress (Junn et al. 2009)
Gene Therapy Based on the Genetic
supporting potential protective effect of this ap-
Causes of PD
proach.
In the last decade, several genes implicated ei- However, a recent study reported that knock-
ther in the Mendelian inheritance of PD or as down of endogenous a-synuclein in the adult rat
risk factors for sporadic PD have been identi- SN leads to the degeneration of nigral dopami-
fied. These findings have dramatically changed nergic neurons and motor deficits (Gorbatyuk
our understanding of the disease process and et al. 2010). Although the mechanisms underly-

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 9


P. Coune et al.

ing this effect remain unclear, it appears that ex- autophagic elimination of dysfunctional mito-
tent of neurodegeneration correlates with the de- chondria (Narendra et al. 2008). Therefore, Par-
gree of a-synuclein silencing. Considering that kin appears as potential neuroprotective agent
the normal function of a-synuclein is still poorly with a broad mode of action, which may con-
understood, the potential consequences of un- tribute to neuronal resistance to various stressors
controlled a-synuclein silencing should be care- potentially implicated in sporadic forms of the
fully addressed, as the near complete loss of a- disease as well.
synuclein may have negative effects on neuronal In rats with a partial unilateral 6-OHDA le-
function and survival. Although challenging, it sion, a lentiviral vector encoding Parkin had a
may be important to devise strategies to geneti- significant neuroprotective effect, leading to a
cally control the degree of a-synuclein silenc- correction of motor asymmetry for 20 weeks
ing to safely implement this approach in PD pa- (Vercammen et al. 2006). Using a more severe
tients. 6-OHDA-induced lesion, a subsequent study
Gene therapy could also provide essential using an AAV vector did not confirm the neuro-
tools for PD cell therapy. Induced pluripotent nal protection, but reported a decrease in drug-
stem cells (iPSC) are stem cells derived from pa- induced rotametry and spontaneous behavior
tients somatic cells that can be differentiated in- in conditions of Parkin overexpression (Man-
to dopaminergic neurons (Soldner et al. 2009). fredsson et al. 2007). It was suggested that Par-
These dopaminergic cells could then be im- kin could improve motor function via increased
planted in the striatum of PD patients to com- levels of tyrosine hydroxylase and striatal dopa-
pensate for the nigral cell loss occurring during mine, thereby enhancing dopamine striatal neu-
PD. However, as these cells are derived from rotransmission. In MPTP-treated mice, an AAV
PD patients, there is a distinct risk that they vector for Parkin expression was reported to in-
www.perspectivesinmedicine.org

could carry mutations perpetuating the pathol- duce significant neuroprotection (Paterna et al.
ogy once grafted in the patients. This issue has 2007).
been addressed in a recent study reporting the Parkin expression has further proved neuro-
generation of iPSC in which disease-causing protective in genetic animal models of PD. In
point mutations have been corrected using zinc Drosophila, Parkin expression has been shown
finger nucleases (Soldner et al. 2011). to reduce dopaminergic neurons degeneration
induced by the Parkin substrate PaelR (Yang
et al. 2003). Studies conducted in rats using
Parkin
lentiviral (Lo Bianco et al. 2004b) and AAV
Parkin has been linked with PD by the discovery (Yamada et al. 2005) vectors showed that Par-
of mutations associated with autosomal reces- kin overexpression significantly reduces the
sive juvenile parkinsonism (AR-JP), an early a-synuclein-induced nigrostriatal degeneration,
onset form of PD with typical symptoms and and leads to behavioral recovery (Yamada et al.
pathology and very slow disease progression 2005). The only primate study conducted yet
(Kitada et al. 1998). Parkin is an E3 ubiquitin did not report a protective effect of Parkin
ligase that polyubiquitylates proteins destined against the a-synuclein-induced loss of dop-
for degradation by the proteasome (Shimura aminergic neurons in the SNpc (Yasuda et al.
et al. 2000). AR-JP mutations lead to partial 2007). However, the study only included two
or complete loss of the protein function (Shi- macaque monkeys and was limited by the only
mura et al. 2000), and therefore to the accu- partial transduction of the dopaminergic nigral
mulation of potentially toxic substrate proteins. neurons achieved using an AAV vector.
In addition to protein ubiquitylation, there is Overall, viral vectors for Parkin expression
also evidence for beneficial effects of Parkin ex- have shown neuroprotective effects in various
pression on oxidative stress levels (Hyun et al. animal models of PD. Targeting the subset of
2002), and more recently, on mitochondrial patients afflicted by AR-JP associated to Parkin
homeostasis, as illustrated by the Parkin-induced deficiency appears as an obvious strategy for the

10 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

clinical application of this etiologic gene ther- parenchyma. With the new recombinant tech-
apy approach. However, despite an early onset, nologies, the vectors are safer and, in the ab-
the Parkin-linked form of the disease has a sence of any viral protein expression, they avoid
very slow progression (Khan et al. 2002), with detrimental host immune reactions.
a good response to L-dopa therapy. These fea- Until now, gene therapy has mainly reached
tures clearly raise the bar for gene therapy ap- clinical trials for the treatment of PD with the
plication. It is therefore important to further rationale of improving the treatments that tar-
explore the function of Parkin and perform get the motor symptoms (Fig. 2). The vectors
more extensive preclinical studies in nonhuman can transfer genetic information to the brain
primates, to determine the possible detrimental and therefore allow for long-term exposure to
effects of long-term Parkin overexpression. factors that would otherwise not cross the
blood-brain barrier. In addition, CNS transgene
delivery is an efficient system to induce the con-
CONCLUDING REMARKS
tinuous expression of enzymes involved in dop-
In the past decade, PD has been the main CNS amine synthesis. By avoiding the fluctuations in
application for gene therapy strategies. This is in brain dopamine concentrations that typically
large part because of the fact that viral vectors lead to side effects in PD, gene therapy offers a
can now efficiently target the neuronal popu- potential alternative to the existing pharmaco-
lations clustered within brain nuclei that are logical treatments. However, as L-dopa therapy
responsible for the major symptoms of PD. In- and DBS already provide significant benefit,
deed, the viral vector technology has made great gene therapy has to prove more effective than
progress. Novel vectors are capable of inducing these treatments to be considered as a reason-
long-term expression in populations of neurons able alternative. Therefore, it is not obvious to
www.perspectivesinmedicine.org

spread over millimeters to centimeters of brain adopt irreversible gene therapy strategies at an

Transgenes Viral vector Preclinical studies Clinical studies

In vitro Rodent Nonhuman


Symptomatic approaches Phase I Phase II Current status
studies models primate

TH + GCH1 + Motor response and reduced


Full ectopic dopamine dyskinesia in rodent and primate
AADC EIAV models
synthesis (Prosavin) Phase I/II clinical trial ongoing

Open Phase I clinical trial: modest


Ectopic L-dopa conversion AADC AAV2 clinical improvements in both on- and
off-states

Motor response and reduced


Ectopic L-dopa synthesis TH + GCH1 AAV5 dyskinesia in rodent models, non-
human primate studies ongoing

Phase II clinical trial: significant


GAD expression in the STN GAD65/67 AAV2 improvement of the UPDRS score
(–8.1 off-medication)

Disease-modifying
approaches

Neurotrophic factors Lentivector/ Phase II clinical trial: no significant


GDNF/Neurturin
improvement of the UPDRS score
gene delivery (CERE-120) AAV2 (AAV2-Neurturin)

Suppression of α-synuclein with viral


Lentivector/ vectors and siRNA infusion
α-Synuclein silencing N/A Potential detrimental effects of
AAV2/5
extensive silencing

Lentivector/ Preclinical studies: neuroprotective


Parkin gene delivery Parkin effect in animal models
AAV2

Figure 2. Current status of the different gene therapy approaches in PD.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 11


P. Coune et al.

early stage of the pathology, in which a signifi- 2011. Bioactivityof AAV2-neurturin gene therapy (CERE-
120): Differences between Parkinson’s disease and non-
cant part of the neurons still survive and the
human primate brains. Movement Disorders 26: 27–36.
therapy has the most chance to succeed. Beck KD, Valverde J, Alexi T, Poulsen K, Moffat B, Vandlen
In the long term, identifying the causes of RA, Rosenthal A, Hefti F. 1995. Mesencephalic dopami-
Parkinson’s disease is a crucial challenge to pre- nergic neurons protected by GDNF from axotomy-in-
vent the disease from progressing to near total duced degeneration in the adult brain. Nature 373:
339 –341.
neuronal loss and untreatable dysfunctions.
Björklund T, Carlsson T, Cederfjäll EA, Carta M, Kirik D.
With the rapid progress in understanding the 2010. Optimized adeno-associated viral vector-mediated
genetic causes of PD, the number of possible striatal DOPA delivery restores sensorimotor function
targets for gene therapy will undoubtedly in- and prevents dyskinesias in a model of advanced Parkin-
son’s disease. Brain 133: 496–511.
crease. In addition, the development of novel
Büning H, Perabo L, Coutelle O, Quadt-Humme S, Hallek
viral vector systems allowing for the induction M. 2008. Recent developments in adeno-associated virus
and fine regulation of transgene expression vector technology. J Gene Med 10: 717 –733.
will provide the needed tools to correct the ge- Carlsson T, Winkler C, Burger C, Muzyczka N, Mandel RJ,
netic defects associated with PD and/or their Cenci A, Björklund A, Kirik D. 2005. Reversal of dyskine-
sias in an animal model of Parkinson’s disease by contin-
downstream toxic effects. Ultimately, such ap- uous L-DOPA delivery using rAAV vectors. Brain 128:
proaches have the potential to curb the rate of 559 –569.
neuronal loss in diseased brain regions. Chen L, Ding Y, Cagniard B, Van Laar AD, Mortimer A, Chi
W, Hastings TG, Kang UJ, Zhuang X. 2008. Unregulated
cytosolic dopamine causes neurodegeneration associated
ACKNOWLEDGMENTS with oxidative stress in mice. J Neurosci 28: 425 –433.
Christine CW, Starr PA, Larson PS, Eberling JL, Jagust WJ,
This work is supported by the Swiss National Hawkins RA, VanBrocklin HF, Wright JF, Bankiewicz
Science Foundation (Grant 31003_120653). KS, Aminoff MJ. 2009. Safety and tolerability of putami-
www.perspectivesinmedicine.org

nal AADC gene therapy for Parkinson disease. Neurology


73: 1662–1669.
Cotzias GC, Papavasiliou PS, Gellene R. 1969. Modification
REFERENCES
of Parkinsonism–chronic treatment with L-dopa. N Engl
Ai Y, Markesbery W, Zhang Z, Grondin R, Elseberry D, Ger- J Med 280: 337–345.
hardt GA, Gash DM. 2003. Intraputamenal infusion of Cunningham LA, Su C. 2002. Astrocyte delivery of glial cell
GDNF in aged rhesus monkeys: Distribution and dopa- line-derived neurotrophic factor in a mouse model of
minergic effects. J Comp Neurol 461: 250 –261. Parkinson’s disease. Exp Neurol 174: 230– 242.
Akerud P, Canals JM, Snyder EY, Arenas E. 2001. Neuropro- Cyr M, Beaulieu J-M, Laakso A, Sotnikova TD, Yao W-D,
tection through delivery of glial cell line-derived neuro- Bohn LM, Gainetdinov RR, Caron MG. 2003. Sustained
trophic factor by neural stem cells in a mouse model of elevation of extracellular dopamine causes motor dys-
Parkinson’s disease. J Neurosci 21: 8108–8118. function and selective degeneration of striatal GABAergic
Azzouz M, Martin-Rendon E, Barber RD, Mitrophanous neurons. Proc Natl Acad Sci 100: 11035– 11040.
KA, Carter EE, Rohll JB, Kingsman SM, Kingsman AJ, Decressac M, Ulusoy A, Mattsson B, Georgievska B, Ro-
Mazarakis ND. 2002. Multicistronic lentiviral vector- mero-Ramos M, Kirik D, Björklund A. 2011. GDNF fails
mediated striatal gene transfer of aromatic L-amino to exert neuroprotection in a rat fag-synuclein model of
acid decarboxylase, tyrosine hydroxylase, and GTP cyclo- Parkinson’s disease. Brain 134: 2302–2311.
hydrolase I induces sustained transgene expression, dop-
Deierborg T, Soulet D, Roybon L, Hall V, Brundin P. 2008.
amine production, and functional improvement in a rat
Emerging restorative treatments for Parkinson’s disease.
model. J Neurosci 22: 10302–10312.
Progress Neurobiol 85: 407– 432.
Bankiewicz KS, Eberling JL, Kohutnicka M, Jagust W, Pivir-
otto P, Bringas J, Cunningham J, Budinger TF, Harvey- Dull T, Zufferey R, Kelly M, Mandel RJ, Nguyen M, Trono D,
White J. 2000. Convection-enhanced delivery of AAV vec- Naldini L. 1998. A third-generation lentivirus vector with
tor in parkinsonian monkeys; in vivo detection of gene a conditional packaging system. J Virol 72: 8463– 8471.
expression and restoration of dopaminergic function us- Eberling JL, Jagust WJ, Christine CW, Starr P, Larson P,
ing pro-drug approach. Exp Neurol 164: 2 –14. Bankiewicz KS, Aminoff MJ. 2008. Results from a phase
Bankiewicz KS, Forsayeth J, Eberling JL, Sanchez-Pernaute I safety trial of hAADC gene therapy for Parkinson dis-
R, Pivirotto P, Bringas J, Herscovitch P, Carson RE, Eckel- ease. Neurology 70: 1980–1983.
man W, Reutter B, et al. 2006. Long-term clinical im- Emborg ME, Carbon M, Holden JE, During MJ, Ma Y, Tang
provement in MPTP-lesioned primates after gene ther- C, Moirano J, Fitzsimons H, Roitberg BZ, Tuccar E, et al.
apy with AAV-hAADC. Mol Ther 14: 564 –570. 2007. Subthalamic glutamic acid decarboxylase gene
Bartus RT, Herzog CD, Chu Y, Wilson A, Brown L, Siffert J, therapy: Changes in motor function and cortical metab-
Johnson EM, Olanow CW, Mufson EJ, Kordower JH. olism. J Cerebral Blood Flow Metabol 27: 501–509.

12 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

Feigin A, Kaplitt MG, Tang C, Lin T, Mattis P, Dhawan V, Ichinose H, Ohye T, Fujita K, Pantucek F, Lange K, Riederer
During MJ, Eidelberg D. 2007. Modulation of metabolic P, Nagatsu T. 1994. Quantification of mRNA of tyrosine
brain networks after subthalamic gene therapy for Par- hydroxylase and aromatic L-amino acid decarboxylase
kinson’s disease. Proc Natl Acad Sci 104: 19559–19564. in the substantia nigra in Parkinson’s disease and schizo-
Fountaine TM, Wade-Martins R. 2007. RNA interference- phrenia. J Neural Transm Park Dis Dement Sect 8: 149–
mediated knockdown of a-synuclein protects human 158.
dopaminergic neuroblastoma cells from MPPþ toxicity Jarraya B, Boulet S, Ralph GS, Jan C, Bonvento G, Azzouz M,
and reduces dopamine transport. J Neurosci Res 85: 351– Miskin JE, Shin M, Delzescaux T, Drouot X, et al. 2009.
363. Dopamine gene therapy for Parkinson’s disease in a non-
Gasmi M, Brandon EP, Herzog CD, Wilson A, Bishop KM, human primate without associated dyskinesia. Sci Transl
Hofer EK, Cunningham JJ, Printz MA, Kordower JH, Med 1: 2ra4.
Bartus RT. 2007a. AAV2-mediated delivery of human Johnston LC, Eberling J, Pivirotto P, Hadaczek P, Federoff
neurturin to the rat nigrostriatal system: Long-term effi- HJ, Forsayeth J, Bankiewicz KS. 2009. Clinically relevant
cacy and tolerability of CERE-120 for Parkinson’s disease. effects of convection-enhanced delivery of AAV2-GDNF
Neurobiol Dis 27: 67– 76. on the dopaminergic nigrostriatal pathway in aged rhesus
Gasmi M, Herzog CD, Brandon EP, Cunningham JJ, Ram- monkeys. Hum Gene Ther 20: 497–510.
irez GA, Ketchum ET, Bartus RT. 2007b. Striatal delivery Junn E, Lee K-W, Jeong BS, Chan TW, Im J-Y, Mouradian
of neurturin by CERE-120, an AAV2 vector for the treat- MM. 2009. Repression of a-synuclein expression and tox-
ment of dopaminergic neuron degeneration in Parkin- icity by microRNA-7. Proc Natl Acad Sci 106: 13052–
son’s disease. Mol Ther 15: 62–68. 13057.
Gill SS, Patel NK, Hotton GR, O’Sullivan K, McCarter R, Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Law-
Bunnage M, Brooks DJ, Svendsen CN, Heywood P. lor PA, Bland RJ, Young D, Strybing K, Eidelberg D, et al.
2003. Direct brain infusion of glial cell line-derived neu- 2007. Safety and tolerability of gene therapy with an
rotrophic factor in Parkinson disease. Nat Med 9: 589 – adeno-associated virus (AAV) borne GAD gene for Par-
595. kinson’s disease: An open label, phase I trial. Lancet
Gorbatyuk OS, Li S, Nash K, Gorbatyuk M, Lewin AS, Sul- 369: 2097– 2105.
livan LF, Mandel RJ, Chen W, Meyers C, Manfredsson FP, Kearns CM, Gash DM. 1995. GDNF protects nigral dopa-
et al. 2010. In vivo RNAi-mediated a-synuclein silencing mine neurons against 6-hydroxydopamine in vivo. Brain
induces nigrostriatal degeneration. Mol Ther 18: 1450– Res 672: 104–111.
www.perspectivesinmedicine.org

1457. Kettler R, Bartholini G, Pletscher A. 1974. In vivo enhance-


Grimm D, Kay MA, Kleinschmidt JA. 2003. Helper virus- ment of tyrosine hydroxylation in rat striatum by tetrahy-
free, optically controllable, and two-plasmid-based pro- drobiopterin. Nature 249: 476–478.
duction of adeno-associated virus vectors of serotypes 1 Khan NL, Brooks DJ, Pavese N, Sweeney MG, Wood NW,
to 6. Mol Ther 7: 839–850. Lees AJ, Piccini P. 2002. Progression of nigrostriatal dys-
Grondin R, Zhang Z, Yi A, Cass WA, Maswood N, Andersen function in a parkin kindred: An [18F]dopa PET and
AH, Elsberry DD, Klein MC, Gerhardt GA, Gash DM. clinical study. Brain 125: 2248–2256.
2002. Chronic, controlled GDNF infusion promotes struc- Kirik D, Rosenblad C, Björklund A. 2000. Preservation of a
tural and functional recovery in advanced parkinsonian functional nigrostriatal dopamine pathway by GDNF in
monkeys. Brain 125: 2191–2201. the intrastriatal 6-OHDA lesion model depends on the
Grondin R, Cass WA, Zhang Z, Stanford JA, Gash DM, site of administration of the trophic factor. Eur J Neurosci
Gerhardt GA. 2003. Glial cell line-derived neurotrophic 12: 3871–3882.
factor increases stimulus-evoked dopamine release and Kirik D, Georgievska B, Burger C, Winkler C, Muzyczka N,
motor speed in aged rhesus monkeys. J Neurosci 23: Mandel RJ, Bjorklund A. 2002. Reversal of motor impair-
1974– 1980. ments in parkinsonian rats by continuous intrastriatal
Hayashita-Kinoh H, Yamada M, Yokota T, Mizuno Y, Mo- delivery of L-dopa using rAAV-mediated gene transfer.
chizuki H. 2006. Down-regulation of a-synuclein expres- Proc Natl Acad Sci 99: 4708–4713.
sion can rescue dopaminergic cells from cell death in the Kish SJ, Tong J, Hornykiewicz O, Rajput A, Chang L-J, Gutt-
substantia nigra of Parkinson’s disease rat model. Bio- man M, Furukawa Y. 2008. Preferential loss of serotonin
chem Biophys Res Commun 341: 1088–1095. markers in caudate versus putamen in Parkinson’s dis-
Hoane MR, Gulwadi AG, Morrison S, Hovanesian G, Lind- ease. Brain 131: 120– 131.
ner MD, Tao W. 1999. Differential in vivo effects of neu- Kishima H, Poyot T, Bloch J, Dauguet J, Condé F, Dollé F,
rturin and glial cell-line-derived neurotrophic factor. Exp Hinnen F, Pralong W, Palfi S, Déglon N, et al. 2004. En-
Neurol 160: 235 –243. capsulated GDNF-producing C2C12 cells for Parkinson’s
Huser D, Gogol-Doring A, Lutter T, Weger S, Winter K, disease: A pre-clinical study in chronic MPTP-treated ba-
Hammer EM, Cathomen T, Reinert K, Heilbronn R. boons. Neurobiol Dis 16: 428– 439.
2010. Integration preferences of wildtype AAV-2 for con- Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y,
sensus rep-binding sites at numerous loci in the human Minoshima S, Yokochi M, Mizuno Y, Shimizu N. 1998.
genome. PLoS Pathogens 6: e1000985. Mutations in the parkin gene cause autosomal recessive
Hyun D-H, Lee M, Hattori N, Kubo S-I, Mizuno Y, Halliwell juvenile parkinsonism. Nature 392: 605–608.
B, Jenner P. 2002. Effect of wild-type or mutant Parkin on Kordower JH, Palfi S, Chen EY, Ma SY, Sendera T, Cochran
oxidative damage, nitric oxide, antioxidant defenses, and EJ, Mufson EJ, Penn R, Goetz CG, Comella CD. 1999.
the proteasome. J Biol Chem 277: 28572–28577. Clinicopathological findings following intraventricular

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 13


P. Coune et al.

glial-derived neurotrophic factor treatment in a patient via enhanced dopamine neurotransmission in a rat
with Parkinson’s disease. Ann Neurol 46: 419– 424. model of Parkinson’s disease. Exp Neurol 207: 289– 301.
Kordower JH, Emborg ME, Bloch J, Ma SY, Chu Y, Leventhal Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioan-
L, McBride J, Chen EY, Palfi S, Roitberg BZ, et al. 2000. nidis JP, Krüger R, Rocca WA, Schneider NK, Lesnick TG,
Neurodegeneration prevented by lentiviral vector deliv- Lincoln SJ, et al. 2006. Collaborative analysis of a-synu-
ery of GDNF in primate models of Parkinson’s disease. clein gene promoter variability and Parkinson disease.
Science 290: 767– 773. JAMA 296: 661–670.
Kordower JH, Herzog CD, Dass B, Bakay RAE, Stansell J, Marks WJ Jr, Ostrem JL, Verhagen L, Starr PA, Larson PS,
Gasmi M, Bartus RT. 2006. Delivery of neurturin by Bakay RA, Taylor R, Cahn-Weiner DA, Stoessl AJ, Ola-
AAV2 (CERE-120)-mediated gene transfer provides struc- now CW. 2008. Safety and tolerability of intraputaminal
tural and functional neuroprotection and neurorestora- delivery of CERE-120 (adeno-associated virus serotype
tion in MPTP-treated monkeys. Ann Neurol 60: 706–715. 2 –neurturin) to patients with idiopathic Parkinson’s
Kumar M, Keller B, Makalou N, Sutton RE. 2001. Systematic disease: An open-label, phase I trial. Lancet Neurol 7:
determination of the packaging limit of lentiviral vectors. 400 –408.
Hum Gene Ther 12: 1893–1905. Marks WJ Jr, Bartus RT, Siffert J, Davis CS, Lozano A, Boulis
Kwon I, Schaffer DV. 2008. Designer gene delivery vectors: N, Vitek J, Stacy M, Turner D, Verhagen L. 2010. Gene de-
Molecular engineering and evolution of adeno-associ- livery of AAV2-neurturin for Parkinson’s disease: A
ated viral vectors for enhanced gene transfer. Pharmaceut double-blind, randomised, controlled trial. Lancet Neu-
Res 25: 489– 499. rol 9: 1164– 1172.
Lee B, Lee H, Nam YR, Oh JH, Cho YH, Chang JW. 2005. McCormack AL, Mak SK, Henderson JM, Bumcrot D, Far-
Enhanced expression of glutamate decarboxylase 65 im- rer MJ, Di Monte DA. 2010. a-synuclein suppression by
proves symptoms of rat parkinsonian models. Gene Ther- targeted small interfering RNA in the primate substantia
apy 12: 1215– 1222. nigra. PloS ONE 5: e12122.
Leriche L, Björklund T, Breysse N, Besret L, Grégoire M-C, Moffat M, Harmon S, Haycock J, O’Malley KL. 1997. L-
Carlsson T, Dollé F, Mandel RJ, Déglon N, Hantraye P, Dopa and dopamine-producing gene cassettes for gene
et al. 2009. Positron emission tomography imaging dem- therapy approaches to Parkinson’s disease. Exp Neurol
onstrates correlation between behavioral recovery and 144: 69–73.
correction of dopamine neurotransmission after gene Montini E, Cesana D, Schmidt M, Sanvito F, Ponzoni M,
www.perspectivesinmedicine.org

therapy. J Neurosci 29: 1544–1553. Bartholomae C, Sergi Sergi L, Benedicenti F, Ambrosi


LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty A, Di Serio C, et al. 2006. Hematopoietic stem cell gene
AW, Eskandar EN, Kostyk SK, Thomas K, Sarkar A, Sid- transfer in a tumor-prone mouse model uncovers low
diqui MS. 2011. AAV2-GAD gene therapy for advanced genotoxicity of lentiviral vector integration. Nat Biotech-
Parkinson’s disease: A double-blind, sham-surgery con- nol 24: 687– 696.
trolled, randomised trial. Lancet Neurol 10: 309–319. Moskalenko M, Chen L, van Roey M, Donahue BA, Snyder
Lim ST, Airavaara M, Harvey BK. 2010. Viral vectors for RO, McArthur JG, Patel SD. 2000. Epitope mapping of
neurotrophic factor delivery: A gene therapy approach human anti-adeno-associated virus Type 2 neutralizing
for neurodegenerative diseases of the CNS. Pharmacol antibodies: Implications for gene therapy and virus struc-
Res 61: 14–26. ture. J Virol 74: 1761–1766.
Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F. 1993. Muramatsu S-I, Fujimoto K-I, Ikeguchi K, Shizuma N, Ka-
GDNF: A glial cell line-derived neurotrophic factor for wasaki K, Ono F, Shen Y, Wang L, Mizukami H, Kume A,
midbrain dopaminergic neurons. Science 260: 1130–1132. et al. 2002. Behavioral recovery in a primate model of
Lo Bianco C, Déglon N, Pralong W, Aebischer P. 2004a. Len- Parkinson’s disease by triple transduction of striatal cells
tiviral nigral delivery of GDNF does not prevent neurode- with adeno-associated viral vectors expressing dopa-
generation in a genetic rat model of Parkinson’s disease. mine-synthesizing enzymes. Hum Gene Ther 13: 345–
Neurobiol Dis 17: 283 –289. 354.
Lo Bianco C, Schneider BL, Bauer M, Sajadi A, Brice A, Iwat- Naldini L, Blömer U, Gage FH, Trono D, Verma IM. 1996a.
subo T, Aebischer P. 2004b. Lentiviral vector delivery of Efficient transfer, integration, and sustained long-term
parkin prevents dopaminergic degeneration in an a-syn- expression of the transgene in adult rat brains injected
uclein rat model of Parkinson’s disease. Proc Natl Acad Sci with a lentiviral vector. Proc Natl Acad Sci 93: 11382–
101: 17510– 17515. 11388.
Luo J, Kaplitt MG, Fitzsimons HL, Zuzga DS, Liu Y, Oshin- Naldini L, Blömer U, Gallay P, Ory D, Mulligan R, Gage FH,
sky ML, During MJ. 2002. Subthalamic GAD gene ther- Verma IM, Trono D. 1996b. In vivo gene delivery and sta-
apy in a Parkinson’s disease rat model. Science 298: ble transduction of nondividing cells by a lentiviral vec-
425–429. tor. Science 272: 263– 267.
Maersch S, Huber A, Büning H, Hallek M, Perabo L. 2010. Narendra D, Tanaka A, Suen D-F, Youle RJ. 2008. Parkin is
Optimization of stealth adeno-associated virus vectors recruited selectively to impaired mitochondria and pro-
by randomization of immunogenic epitopes. Virology motes their autophagy. J Cell Biol 183: 795–803.
397: 167– 175. Nishioka K, Hayashi S, Farrer MJ, Singleton AB, Yoshino H,
Manfredsson FP, Burger C, Sullivan LF, Muzyczka N, Lewin Imai H, Kitami T, Sato K, Kuroda R, Tomiyama H, et al.
AS, Mandel RJ. 2007. rAAV-mediated nigral human par- 2006. Clinical heterogeneity of a-synuclein gene duplica-
kin over-expression partially ameliorates motor deficits tion in Parkinson’s disease. Ann Neurol 59: 298– 309.

14 Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431


Parkinson’s Disease: Gene Therapies

Nutt JG, Burchiel KJ, Comella CL, Jankovic J, Lang AE, Laws minergic neurons following striatal 6-hydroxydopamine
ER, Lozano AM, Penn RD, Simpson RK, Stacy M, et al. lesion. Proc Natl Acad Sci 92: 8935– 8939.
2003. Randomized, double-blind trial of glial cell line- Shen Y, Muramatsu SI, Ikeguchi K, Fujimoto KI, Fan DS,
derived neurotrophic factor (GDNF) in PD. Neurology Ogawa M, Mizukami H, Urabe M, Kume A, Nagatsu I,
60: 69– 73. et al. 2000. Triple transduction with adeno-associated vi-
Olsen JC. 1998. Gene transfer vectors derived from equine rus vectors expressing tyrosine hydroxylase, aromatic-
infectious anemia virus. Gene Therapy 5: 1481– 1487. L-amino-acid decarboxylase, and GTP cyclohydrolase I
Palfi S, Leventhal L, Chu Y, Ma SY, Emborg M, Bakay R, for gene therapy of Parkinson’s disease. Hum Gene Ther
Déglon N, Hantraye P, Aebischer P, Kordower JH. 2002. 11: 1509–1519.
Lentivirally delivered glial cell line-derived neurotrophic Shimura H, Hattori N, Kubo Si, Mizuno Y, Asakawa S, Min-
factor increases the number of striatal dopaminergic neu- oshima S, Shimizu N, Iwai K, Chiba T, Tanaka K, et al.
rons in primate models of nigrostriatal degeneration. J 2000. Familial Parkinson disease gene product, parkin,
Neurosci 22: 4942– 4954. is a ubiquitin-protein ligase. Nat Gen 25: 302–305.
Park K. 2001. Protection of nigral neurons by GDNF-engi- Singleton AB, Farrer M, Johnson J, Singleton A, Hague S,
neered marrow cell transplantation. Neurosci Res 40: Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nuss-
315–323. baum R, et al. 2003. a-Synuclein locus triplication causes
Pascual A, Hidalgo-Figueroa M, Piruat JI, Pintado CO, Parkinson’s disease. Science 302: 841.
Gómez-Dı́az R, López-Barneo J. 2008. Absolute require- Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook
ment of GDNF for adult catecholaminergic neuron sur- EG, Hargus G, Blak A, Cooper O, Mitalipova M, et al.
vival. Nat Neurosci 11: 755–761. 2009. Parkinson’s disease patient-derived induced pluri-
Paterna J-C, Feldon J, Büeler H. 2004. Transduction profiles potent stem cells free of viral reprogramming factors. Cell
of recombinant adeno-associated virus vectors derived 136: 964–977.
from serotypes 2 and 5 in the nigrostriatal system of Soldner F, Laganière J, Cheng AW, Hockemeyer D, Gao Q,
rats. J Virol 78: 6808– 6817. Alagappan R, Khurana V, Golbe LI, Myers RH, Lindquist
Paterna J-C, Leng A, Weber E, Feldon J, Büeler H. 2007. DJ-1 S, et al. 2011. Generation of isogenic pluripotent stem
and Parkin modulate dopamine-dependent behavior and cells differing exclusively at two early onset Parkinson
inhibit MPTP-induced nigral dopamine neuron loss in point mutations. Cell 146: 318–331.
mice. Mol Ther 15: 698–704. Sun M, Zhang G-R, Kong L, Holmes C, Wang X, Zhang W,
Goldstein DS, Geller AI. 2003. Correction of a rat model
www.perspectivesinmedicine.org

Rahim AA, Wong AMS, Howe SJ, Buckley SMK, Acosta-


Saltos AD, Elston KE, Ward NJ, Philpott NJ, Cooper of Parkinson’s disease by coexpression of tyrosine hy-
JD, Anderson PN, et al. 2009. Efficient gene delivery to droxylase and aromatic amino acid decarboxylase from
the adult and fetal CNS using pseudotyped non- a helper virus-free herpes simplex virus type 1 vector.
integrating lentiviral vectors. Gene Therapy 16: 509 –520. Hum Gene Ther 14: 415– 424.
Rosenblad C, Kirik D, Devaux B, Moffat B, Phillips HS, Tseng JL, Baetge EE, Zurn AD, Aebischer P. 1997. GDNF
Björklund A. 1999. Protection and regeneration of nigral reduces drug-induced rotational behavior after medial
dopaminergic neurons by neurturin or GDNF in a partial forebrain bundle transection by a mechanism not involv-
lesion model of Parkinson’s disease after administration ing striatal dopamine. J Neurosci 17: 325 –333.
into the striatum or the lateral ventricle. Eur J Neurosci Vercammen L, Van der Perren A, Vaudano E, Gijsbers R, De-
11: 1554–1566. byser Z, Van den Haute C, Baekelandt V. 2006. Parkin
Sajadi A, Bensadoun J-C, Schneider BL, Lo Bianco C, Ae- protects against neurotoxicity in the 6-hydroxydopamine
bischer P. 2006. Transient striatal delivery of GDNF via rat model for Parkinson’s disease. Mol Ther 14: 716– 723.
encapsulated cells leads to sustained behavioral improve- Yamada M, Mizuno Y, Mochizuki H. 2005. Parkin gene ther-
ment in a bilateral model of Parkinson disease. Neurobiol apy for a-synucleinopathy: A rat model of Parkinson’s
Disease 22: 119 –129. disease. Hum Gene Ther 16: 262–270.
Samulski RJ, Chang LS, Shenk T. 1989. Helper-free stocks of Yang Y, Nishimura I, Imai Y, Takahashi R, Lu B. 2003. Parkin
recombinant adeno-associated viruses: Normal integra- suppresses dopaminergic neuron-selective neurotoxicity
tion does not require viral gene expression. J Virol 63: induced by Pael-R in Drosophila. Neuron 37: 911– 924.
3822– 3828. Yasuda T, Miyachi S, Kitagawa R, Wada K, Nihira T, Ren Y-R,
Samulski RJ, Zhu X, Xiao X, Brook JD, Housman DE, Hirai Y, Ageyama N, Terao K, Shimada T, et al. 2007.
Epstein N, Hunter LA. 1991. Targeted integration of ad- Neuronal specificity of a-synuclein toxicity and effect
eno-associated virus (AAV) into human chromosome 19. of Parkin co-expression in primates. Neuroscience 144:
EMBO J 10: 3941– 3950. 743 –753.
Sapru MK, Yates JW, Hogan S, Jiang L, Halter J, Bohn MC. Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. 1997.
2006. Silencing of human a-synuclein in vitro and in rat Multiply attenuated lentiviral vector achieves efficient
brain using lentiviral-mediated RNAi. Exp Neurol 198: gene delivery in vivo. Nat Biotechnol 15: 871 –875.
382–390. Zufferey R, Dull T, Mandel RJ, Bukovsky A, Quiroz D, Nal-
Sauer H, Rosenblad C, Björklund A. 1995. Glial cell line- dini L, Trono D. 1998. Self-inactivating lentivirus vector
derived neurotrophic factor but not transforming growth for safe and efficient in vivo gene delivery. J Virol 72:
factor b 3 prevents delayed degeneration of nigral dopa- 9873–9880.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a009431 15

You might also like