REVIEW
Potential cellular and regenerative approaches
for the treatment of Parkinson’s disease
Emma L Lane
Olivia J Handley
Anne E Rosser
Stephen B Dunnett
Brain Repair Group, School
of Biosciences, Cardiff University,
CF10 3US, UK
Correspondence: Emma Lane
Brain Repair Group, School
of Biosciences, Cardiff University,
Cardiff, CF10 3US, UK
Tel +44 29 2087 4112
Fax +44 29 2087 6749
Email laneel@cf.ac.uk
Abstract: Parkinson’s disease is most commonly treated with a range of pharmacotherapeutics,
with the more recent introduction of surgical techniques including deep-brain stimulation.
These have limited capabilities to improve symptoms of the disease in more advanced stages,
thus new therapeutic strategies including the use of viral vectors and stem cells are in development. Providing a continuous supply of dopamine to the striatum in an attempt to improve
the treatment of motor symptoms using enzymes in the dopamine synthesis and machinery is
one approach. Alternatively, there are tools which may serve to both protect and encourage
outgrowth of surviving neurons using growth factors or to directly replace lost innervation by
transplantation of primary tissue or stem cell-derived dopaminergic neurons. We summarize
some of the potential therapeutic approaches and also consider the recent EU directives on
practical aspects of handling viral vectors, cells and tissues, and in the running of clinical trials
in Europe which impact on their development.
Keywords: transplantation, viral vector, stem cells, ethics, European Union directive
Parkinson’s disease (PD) is one of the most common neurodegenerative diseases
affecting around 1%–2% of the population over 70 years of age (MacDonald et al
2000). Motor symptoms are typically first observed as tremor in one extremity; the
disease then continues its relentless progression throughout the body. It causes not only
the cardinal symptoms of resting tremor, postural instability, rigidity, bradykinesia,
and akinesia but also many other symptoms including autonomic dysfunction, depression, and sleep disturbances which can present prior to the motor disorder (Chaudhuri
et al 2006). Many of the motor symptoms are ascribed to the specific deterioration of
dopaminergic neurons of the nigrostriatal pathway that typifies the disease. Pigmented
cell bodies residing in the A9 substantia nigra region of the hindbrain with heavily
branched projections targeted mainly at the putamen degenerate retrogradely, striatal
innervation being lost first, followed by cell body deterioration. Additional pathology in
specific neurochemical systems is observed in the loss of serotonergic, noradrenergic,
and cholinergic cells. The pathological criteria for diagnosis are cellular protein inclusions termed Lewy bodies (Jellinger 1991). The extent of these varies enormously
from patient to patient and may include areas outside of the nigrostriatal system, for
example, the cortex.
The primary therapeutic antiparkinsonian approach in the clinic at present is
pharmaceutics-based. Symptomatic treatments are highly effective at managing the
motor consequences of nigrostriatal loss, especially in the early stages. However, there is
currently no conclusive evidence that any drug licensed for PD has the ability to prevent
the relentless wave of degeneration the disease typifies. Ergot and non-ergot derived
dopamine D2 receptor agonists and L-dopa, the metabolic precursor to dopamine
Neuropsychiatric Disease and Treatment 2008:4(5) 835–845
© 2008 Dove Medical Press Limited. All rights reserved
835
Lane et al
(with a peripheral decarboxylase inhibitor), constitute the
mainstay therapies, although there are also a number of
adjunctive therapies in use. Over recent years, there has
been increasing interest in surgical approaches, which are
now in routine use for specific symptoms in many countries.
Previously ablative surgeries such as pallidotomy or subthalamotomy have been successfully performed and found
to improve certain aspects of motor function. However, this
has generally now been replaced by the more easily refined
approach of deep-brain stimulation (DBS), involving high
frequency stimulation of specific nuclei, most commonly
the subthalamic nucleus. While generally effective, these
current treatments also have significant inadequacies. The
long term use of drug treatments produces multiple sideeffects, not least the ‘wearing off’ phenomena in which the
efficacy of the drug wanes, giving way to the development
of motor fluctuation and the onset of abnormal involuntary
movements known collectively as ‘dyskinesia’. As the disease
progresses, ever-increasing doses of L-dopa are required to
control the primary symptoms, which exacerbates the side
effects, eventually limiting treatment options. There are also
symptoms that are non-responsive to drug treatment, such as
postural instability and dementia. When applied appropriately
DBS can be highly effective at relieving some motor symptoms and reducing the need for L-dopa, but it is costly and
generally only considered for patients with late stage disease
and without signs of dementia. Furthermore, there is concern
about significant side effects such as cognitive decline, which
may be permanent.
As a result of these inadequacies, alternative and supplementary symptomatic therapies are still being sought and
a number of different pharmacotherapeutic avenues are
being explored. These are reviewed in more detail elsewhere (see Schapira et al 2006) but briefly include other
potential drugs targeting the dopaminergic system such as
dopamine-modulating agents (Ekesbo et al 2000; Pirker
et al 2001) and monoamine uptake inhibitors (Frackiewicz
et al 2002; Bara-Jimenez et al 2004). Furthermore, drugs
affecting the serotonin system, such as the 5-HT1A agonist
sarotizan (Bara-Jimenez et al 2005; Goetz et al 2007);
adenosine A2A antagonists; opioids; and glutamatergic and
GABAergic drugs are at different stages of development.
These compounds can act at several levels: either acting to
directly replace the lost dopaminergic transmission at the
level of the striatum; acting through alternate mechanisms
in the striatum to effect striatal output; or targeting downstream nuclei in the basal ganglia to redress the imbalance
in motor output signals.
836
Some of the aforementioned symptomatic therapies
are purported to have neuroprotective potential on the
remaining dopaminergic neurones. The rate of progression
of the disease on patients taking the selective irreversible
monoamine oxidase inhibitors selegiline is slower than
those on L-dopa alone suggesting that there is a supplementary action in slowing the degenerative process (Shoulson
1998). However, the data are ambiguous and the design of
the clinical trials cannot rule out an entirely symptomatic
effect. Multiple mechanisms have been proposed to be
involved in the degenerative process, many overlapping with
other neurodegenerative diseases. Thus, anti-oxidants, like
coenzyme Q10 and vitamins C and E, are being evaluated
in different disease conditions for their ability to slow down
the disease process.
With the exception of targeted surgical intervention all
of the above are systemic therapies, however a generation
of potential therapies are under development, which involve
more directed cellular approaches to the treatment of PD.
In this review, we examine the different methodological
approaches to deliver symptomatic, neuroprotective and
neurorestorative agents directly to the affected areas of
the brain.
The focus of degeneration in PD is the nigrostriatal
dopaminergic loss, with the greatest impact being in the
loss of input to the striatum. Rebuilding the nigrostriatal
tract in an adult in the absence of developmental signals and
specific growth signals, has proved highly complex and thus
restorative approaches are predominantly focused on placing
dopamine cells into the nigrostriatal dopamine neuron target
site. Methodological approaches include the use of viral
vectors, either directly administered or used to alter cell
function prior to transplantation. Furthermore, donor cells
can either be transplanted and allowed to integrate with the
host environment or encapsulated to facilitate removal. Their
development has not only entered novel scientific arenas but
has raised practical and ethical concerns which have been
addressed at international level with the introduction of new
EU legislation to govern these processes. We focus on the
areas of this legislation that has greatest practical impact
on researchers throughout Europe in the context of these
developing therapies.
Symptomatic strategies
The degeneration of dopaminergic neurons in PD causes the
striatum to be depleted of not only dopamine, but also the
dopamine synthesizing machinery that is present in terminal
axonal regions. There is great interest therefore, in enhancing
Neuropsychiatric Disease and Treatment 2008:4(5)
Treatments for PD and current EU legislation
either L-dopa or dopamine production by equipping other
cells in the striatum with the necessary ‘tools’ through the use
of viral vectors (Carlsson et al 2007). L-dopa is converted to
dopamine either by the remaining dopaminergic terminals or
by serotonergic terminals or glia in the striatum (Tanaka et al
1999; Maeda et al 2005). The production of dopamine from
L-tyrosine is a multistage process, the rate-limiting enzyme
being tyrosine hydroxylase, which converts L-tyrosine to
the dopamine precursor L-dopa. However, supplementation
with additional tyrosine hydroxylase alone is not a viable
approach as its enzymatic functioning requires the presence of a co-factor tetrahydrobiopterin (BH4) (Fukushima
et al 1977). Hence, in order to produce functional benefit,
a combination of viral vectors expressing both GTP cyclohydrolase 1 (GCH1; a key enzyme in BH4 synthesis) and
tyrosine hydroxylase is required. The advantage of the use
of viral vectors in this context is the continuous delivery of
dopamine. Continuous enteral infusion of L-dopa or subcutaneous apomorphine significantly reduces motor fluctuations
and dyskinesia demonstrating that the pulsatile changes in
dopamine levels after oral L-dopa administration may be
responsible for their generation (Sage and Mark 1992; Syed
et al 1998; Poewe and Wenning 2000; Nyholm et al 2003).
Viral vector enhanced production of L-dopa should produce
a constant level of dopamine to provide good therapeutic
benefit but without the development of dyskinesia. Results
in the 6-OHDA lesioned rat demonstrate robust behavioral
recovery as assessed by amphetamine-induced rotation and
the cylinder test of forelimb asymmetry concurrent with
a reduction in L-dopa induced dyskinesia (Carlsson et al
2005). L-dopa delivered to the periphery is subjected to
premature conversion to dopamine that necessitates the
co-administration of a peripheral amino acid decarboxylase
inhibitor (AADC) to increase central uptake of L-dopa.
Viral vector mediated enhancement of dopamine synthesis
in the striatum would make the use of such supplementary
oral drugs redundant. Conversely, it has inspired the use
of a triple viral approach delivering AADC intrastriatally
in addition to TH and GCH1 to facilitate the conversion of
L-dopa to dopamine (Sun et al 2004). This approach, using
both AAV and lentiviral vectors, improves motor function
in rats and primates and increases dopamine levels in the
affected striatum (Muramatsu et al 2002).
A potentially simpler approach is to transduce with a viral
vector delivering AADC alone (Leff et al 1999). As AADC
is lost concomitantly with the degenerating dopaminergic
neurons, increasing conversion of L-dopa to dopamine will
maximize the efficacy of the absorbed L-dopa. However,
Neuropsychiatric Disease and Treatment 2008:4(5)
this apparently more simple approach could be more risky
as there is no inherent feedback mechanism in the control of
AADC activity and increased concentrations of AADC could
lead to dopamine over production. While TH is inhibited by
dopamine, self-limiting dopamine production, AADC has
no such innate control mechanism (Carlsson et al 2007).
This could lead to even greater degrees of fluctuating striatal
dopamine and exacerbate motor disturbances.
One consequence of dopaminergic input in the striatum
is disinhibition of the subthalamic nucleus. This is the most
common target of DBS, in which high frequency stimulation acts to reduce the activity of this nucleus. Similarly,
transfection with an AAV encoding the gene for glutamic
acid decarboxylase, synthesizing the inhibitory transmitter
GABA has been shown to reduce the excitatory output from
the subthalamic nucleus, improving motor cortex activity
and consequently in both rodent and macaques models of
PD restoring motor function (Luo et al 2002; Lee et al 2005;
Emborg et al 2007).
Neurotrophic factors
In the region of 70%–80% of striatal dopamine is depleted
before significant motor symptoms are detected in patients
(Bernheimer et al 1973). This suggests that there is a significant pre-symptomatic interval during which some degree of
cellular plasticity allows the brain to compensate for the lost
innervation. Sprouting of remaining dopaminergic fibres after
neurotoxic challenge has been reported in 6-OHDA lesion
models of PD, and changes in presynaptic terminals, upregulation of AADC levels to increase dopamine conversions and
reduced dopamine transporter have been demonstrated both
in MPTP primate models and PD patients. These compensatory mechanisms have the potential to be harnessed to further
delay the onset of symptoms or prevent deterioration.
Neurotrophic factors are a family of protein growth
factors that regulate components of neuronal growth. Neuron
number, branching, axon and dendrite length, neuronal phenotype and synaptic plasticity can all be regulated through
their action at specific receptors. Due to the differential
expression of these growth factors and their receptors, different neuronal populations are influenced to a greater or lesser
extent by each factor. GDNF, neurturin, CNFT and BDNF,
to name but a few, have all been evaluated in animal models
of PD to determine their effects on dopaminergic neurons.
Many molecules have been shown to be neuroprotective,
reducing dopaminergic cell death following toxic challenges.
GDNF has been the most potent trophic molecule used so
far and in animal models of PD has clearly demonstrated a
837
Lane et al
capacity to protect neurons against a toxic challenge (Zurn
et al 2001; Sherer et al 2006; Patel and Gill 2007). However,
these factors require application directly into the relevant
area as they do not cross the blood-brain barrier and even
when delivered directly into the brain, the precise location
of the cannula seems critical. The first studies used intracerebroventricular pumps containing GDNF, which failed
to improve symptoms and were associated with serious side
effects likely to be as a result of the extrastriatal delivery
(Kordower et al 1999; Nutt et al 2003).
The first successful application of GDNF was in the form
of a pump installed in the abdomen that released GDNF
into the putamen via a thin cannula. The first intraputaminal phase I open label safety trial on 5 patients produced
dramatic improvements in motor function in all of the patients
(Gill et al 2003). However, a double-blind placebo-controlled
trial sponsored by Amgen designed to further examine
the benefits of this approach was prematurely terminated
(Lang et al 2006). The primary outcome measures had not
been achieved and anti-GDNF antibodies had been found in
some patients indicating a possible autoimmune response.
Furthermore, very high doses of GDNF administered to
primates had produced cerebellar lesions. Various technicalities have been given for the failure of this trial including the
efficiency of delivery due to use of different cannulae from
the initial trial (Lang et al 2006).
Since the potential of GDNF has been determined, there
have been alternative approaches to the delivery of the
growth factor. The use of viral vectors to engineer cells to
produce GDNF within the striatum is being considered to
produce a continuous supply, rather than the re-fills required
by a pump. The viral vector could be injected directly in
vivo transfecting the host striatal cells and enabling them
to express GDNF. In 6-OHDA lesioned rats, transfection
of adeno-associated viral vectors expressing the GDNF
protein have significantly protected dopaminergic terminals in the striatum from insult by 6-OHDA in both rodents
and non-human primates (Choi-Lundberg et al 1998; Kirik
et al 2000; Wang et al 2002; Eslamboli et al 2003). Problems with this approach include control of the expression
of the vector should there be side effects. It has already
been demonstrated that aberrant sprouting of fibres and
downregulation of tyrosine hydroxylase expression occurs
(Georgievska et al 2002; Rosenblad et al 2003) which
could potentially limit functional recovery in the long term.
Other possible approaches are to transfect cells in vitro and
either transplant these GDNF producing cells directly or,
more safely, deliver encapsulated cells in a semi-permeable
838
membrane. Such encapulsated GDNF-producing cells have
protected nigrostriatal dopaminergic neurons from 6-OHDA
toxicity (Shingo et al 2002; Yasuhara et al 2005) and also
shown a neurorestorative action by increasing dopamine
fibre density, an effect that was sustained following capsule
removal (Sajadi et al 2006). The advantages to this approach
are that multiple devices could be used to provide GDNF
throughout the putamenal area, the lower levels delivered in
this approach should minimize side effects and the risks of
cerebellar toxicity and antibody generation highlighted by
the Amgen trial. As with direct viral vector administration,
long-term safety trials firstly in non-human primates need to
be carried out to assess the ease of implantation and retrieval,
efficacy and also toxicity and immunogenicity (Lindvall and
Wahlberg 2008). One disadvantage of this approach concerns
the fact that capsules have a limited lifespan and therefore
surgery would need to be repeated.
Other growth factors of the same family, neurturin and
CDNF, have also been proposed as potential therapeutics.
Both growth factors protect dopaminergic neurons from toxic
insult in vivo (Rosenblad et al 1999; Li et al 2003; Lindholm
et al 2007) but while CDNF is still in the early stages of
preclinical studies, neurturin has progressed to clinical trials using intrastriatal delivery through an adeno-associated
virus type II (AAV2). Preliminary safety and efficacy data
from the phase I clinical trial of 12 patients are encouraging
with no significant side effects and an improvement of up
to 36% in the UPDRS score in 9 of the 12 patients (Marks
2006) and phase II trials are underway.
Cell transplantation
Transplantation of primary ventral mesencephalic tissue
into the striatum aims to restore brain circuitry and function
lost as a result of PD. The main objective of primary tissue
transplantation has been to provide proof-of-principle that
grafted dopaminergic neurons can i) survive and restore
regulated dopamine release, ii) integrate with the host brain
to reinstate frontal cortical connections and activation,
and iii) lead to measurable clinical benefits together with
improved quality of life.
Preclinical work in animal models of PD has shown that
grafted dopaminergic neurons, extracted from the developing
ventral mesencephalon (VM) can survive, reinnervate the
lesioned striatum, and improve motor function (Bjorklund
1992; Herman and Abrous 1994; Winkler et al 2000). Over
the past two decades, a series of open-label clinical trials have
provided convincing evidence to show that human embryonic
nigral neurons taken at a stage of development when they
Neuropsychiatric Disease and Treatment 2008:4(5)
Treatments for PD and current EU legislation
are committed to a dopaminergic phenotype can survive,
integrate and function over a long time in the human brain. To
date, approximately 350 patients with PD world-wide have
received primary tissue transplantation. Based on a small
number of carefully controlled clinical trials, there is good
evidence of graft survival, with grafted neurons developing
afferent and efferent projections with the host neurons. Longterm survival of dopaminergic grafts is possible up to 10 years
after transplantation (Piccini et al 1999), and there have been
no reported cases of overt immunorejection even after several
years of withdrawal from immunosuppression (Olanow et al
2003). Evidence from PET scanning has revealed significant
increases in activation in the areas reinnervated by the grafted
cells, and longitudinal clinical assessments indicate significant functional recovery for motor control, in some cases for
more than 10 years (Dunnett et al 2001; Lindvall and Hagell
2002; Bjorklund et al 2003). In the most successful cases,
patients have either reduced dependency for or completely
withdrawn from L-dopa treatment. Post-mortem studies
similarly show good survival of transplanted neurons and
well integrated grafts (Kordower et al 1995).
Two NIH sponsored double-blind placebo-controlled
clinical trials have been unable to replicate the scale of
these clinical benefits and reported severe graft-induced
dyskinesias during “off” phases (Freed et al 2001; Olanow
et al 2003). The precise mechanism responsible for these
dyskinesias remains unknown but it does not appear to be
related to graft overgrowth resulting in excessive dopamine
release (Hagell et al 2002; Lane et al 2006). One possibility
surrounds the quality of dissected tissue. Successful trials
have used either freshly dissected tissue or tissue that has
been stored in culture for only a few days. One of the trials
reporting cases of severe dyskinesias used tissue stored in
culture for up to four weeks (Freed et al 2001) and it may
be that holding tissue in this way reduces its dopaminergic
composition (Hagell et al 2002). A further issue concerns
the identification of dense hyperdopaminergic areas within
the graft of some patients with graft-induced dyskinesias
(Ma et al 2002). This may have caused uneven striatal innervation and excessive dopamine release into non-reinnervated
areas (Winkler et al 2005). Finally, it should be noted that
these studies were severely under-powered, and moreover,
subsequent follow-up data have suggested some evidence
of efficacy.
It is also possible that variable side effects of graftinduced dyskinesias are related to patient selection. Greater
functional improvement is associated with younger patients,
and in patients with less advanced disease. This is most
Neuropsychiatric Disease and Treatment 2008:4(5)
likely because the neuropathology is relatively confined
to the nigro-striatal pathway and may have better trophic
support compared to patients with more advanced disease.
Furthermore, patients with more advanced disease and who
have become dyskinetic from long-term use of L-dopa may
have increased susceptibility to developing graft-induced
dyskinesias (Piccini et al 2005; Lane et al 2006).
VM grafts have thus far been placed in the putamen, and
patients who received most benefit from these transplants
have had pathology restricted to this area both prior to and
after transplantation (Piccini et al 2005). Given the range
of symptoms and pathology with different levels of severity seen in PD patients it has been proposed that placement
of VM grafts should be based on patient-specific profiles
determined by FD-PET imaging (Winker et al 2005). The
findings observed in the NIH sponsored trials have led to
stimulating and extensive review of the different methodological approaches used by different centers to procure,
handle and store fetal tissue. It is possible that differences
between these approaches may account for the variability
in graft-induced functional recovery. A recent review by
Winkler et al (2005) identifies the following aspects requiring harmonisation ahead of further clinical trials: dissection
of the tissue, the age of donor foetus, the length and type
of storage after dissection, the method of dissociation prior
to grafting (pieces vs suspension), and the composition of
medium used for storage and/or implantation. The limited
availability of tissue and the need for multiple donors for
single patients leads to problems of co-ordinating and storing tissue prior to transplantation. The quality of the tissue
is intrinsically variable and reaching consistent standards of
reproducible preparation, quality control and safety assessment to compare accurately the safety and efficacy of this
therapy across different trials are very difficult to achieve
through this source of tissue. Therefore, while the use of
primary tissue has allowed the proof-of-principal studies to
be completed, permitting the accumulation of a substantial
body of experience, they have also reinforced the fact that
alternate source of cells are necessary for transplantation to
become a viable therapeutic option.
Stem cells
Stem cells could provide one such source and would overcome the issue of limited availability of fresh primary fetal
cells. A wide range of stem cells are being investigated as
potential sources of dopaminergic neurons for transplantation
and while it is beyond the scope of this review to go through
each of these in detail we will give a short update as to the
839
Lane et al
progress being made (for more detailed review see Dass et al
2006; Morizane et al 2008; Zietlow et al 2008). Stem cells
can be obtained from various sources including those derived
from the early developing embryo (embryonic stem cells),
or later in development from gonadal regions (embryonic
germ cells), from the developing or adult brain (neural stem
cells) and other tissues such as bone marrow, umbilical
cord and amniotic fluid and very recently, the carotid body
(mesenchymal stem cells). The majority of research thus far
with respect to the formation of dopaminergic neurons for
the treatment of PD is in embryonic stem cells and neural
stem cells. Dopaminergic neurons are more easily obtained
from neural stem cells in the developing VM than other parts
of the developing central nervous system but the number
of dopaminergic cells produced is still very low. Despite
genetic manipulation and the addition of various growth
and differentiation factors, generating large numbers of
dopaminergic cells from this cell type has had mixed results
(Wang et al 2004; Yang et al 2004). However, greater success
has been achieved with the more complex embryonic stem
cells. Derived from blastocysts donated following in vitro
fertilization these cells are truly pluripotent. Promising data
have been obtained with dopaminergic neurons derived from
mouse ES cells, significantly improving motor function in
a rat model of PD (Kim et al 2002). However, directing the
differentiation of human ES cells has proved complex and
while 50% of cells spontaneously differentiate into neurons
upon LIF withdrawal, few are dopaminergic. Thus, there is
the need to develop protocols to ‘direct’ differentiation. The
most successful published protocols describe multiple culture
stages in which different transcription and growth factors are
added at controlled time points (Perrier et al 2004; Roy et al
2006). However, despite good yield of dopaminergic neurons
in vitro, clinically relevant long-term survival and behavioral
recovery in animal models rivalling that of primary tissue has
yet to be convincingly demonstrated (Ben-Hur et al 2004;
Park et al 2005; Brederlau et al 2006; Roy et al 2006). The
use of embryonic stem cells is still ethically controversial
and there is a risk of unregulated growth into undesirable cell
types or tumor formation. Neuronal stem cells carry less risk
in this respect but furthermore, unlike embryonic stem cells,
which are only derived from the embryonic blastocyst, neural
stem cells can be found both in embryonic neural tissue and
also in specific neurogenic regions of the adult brain. If the in
vivo survival of neural stem cells can be improved they hold
the potential to provide autologous transplantation as patients
provide the cells for their own recovery. Cell lines that also
hold that potential and that could be obtained through less
840
invasive means are carotid body and bone marrow derived
mesenchymal stem cells.
Interestingly, stem cells may not just be useful as
dopamine factories in the striatum. Recent studies in both
rodent and primate models have shown significant behavioral recovery following transplantation with neural stem
cells. In addition to the generation of a small population
of dopaminergic neurons other cells within the graft were
found to be releasing growth factors which are purported
to exert neuroprotective or neuroregenerative influences
(Redmond et al 2007; Yasuhara et al 2007). While more
evidence needs to be accumulated on the longevity of this
effect, it broadens the potential of neural stem cells from
simple dopamine replacement to preserving and enhancing
remaining dopaminergic neurons.
Neurogenesis
As mentioned above endogenous stem cells are present
in specific regions of the brain. While the occurrence of
neurogenesis in the striatum and substantia nigra is debated
(Zhao et al 2003; Frielingsdorf et al 2004), one indisputable
neurogenic region is the subventricular zone (SVZ) lying
adjacent to the striatum. The cells in the region are an assortment of stem and progenitor cells that have the potential to
be mobilized and induced to differentiate by the presence
of growth factors or other small molecules. In the normal
condition 75%–99% or the cells differentiate into granular
GABAergic neurons, with the rest forming periglomular
neurons expressing either tyrosine hydroxylase or GABA.
The control of proliferation and mobilisation of these cells
may be dopaminergic as both MPTP and 6-OHDA mediated
dopamine depletion reportedly decrease proliferation in this
zone (Baker et al 2004; Hoglinger et al 2004). An additional
source of endogenous source of new dopaminergic neurons
may be in the recently described presence of tyrosine hydroxylase positive cell bodies in the striatum, which increase in
quantity with dopaminergic denervation (Dubach et al 1987;
Betarbet et al 1997; Porritt et al 2000). This is believed to
be a phenotypic switch in striatal neurons in response to
low dopamine levels (Tande et al 2006). As yet there are no
imminent therapeutic strategies heading towards the clinic
that manipulate these endogenous systems but their potential
is waiting to be harnessed. Therapeutic strategies to increase
striatal dopamine could involve recruiting newly produced
neurons in the SVZ and encouraging them to migrate into
the striatum and differentiate into dopaminergic neurones or
to stimulate cells resident in the striatum. In order for this to
be achieved understanding more about these two processes
Neuropsychiatric Disease and Treatment 2008:4(5)
Treatments for PD and current EU legislation
of neurogenesis and phenotypic switching in the striatum
is necessary, determining the intrinsic or extrinsic factors
responsible may provide an alternative set of mechanisms
that could be utilized to treat PD.
Clinical trials of new therapies
Clinical trials of restorative therapy in PD must follow
clinical and research governance frameworks to protect
and promote the health and well-being of the patient at all
times. Successful frameworks are structured around three
main aims: establishing clear lines of responsibility and
accountability, implementing the highest possible standards of clinical care, and promoting a constant dynamic of
improvement. Within the European Union (EU), legislative
acts known as EU directives, require EU member states to
achieve a particular result without laying down the means of
accomplishing that result, leaving the individual members
to devise their own advisory and enforcement bodies. Here
we describe two EU directives that need to be considered in
clinical trials of restorative therapies for PD, whether of cell
transplantation or gene therapy.
EU directive 2001/20/EC relating
to the implementation of good
clinical practice in the conduct
of clinical trials on medicinal
products for human use
The purpose and scope of this directive is to enhance the
protection of human subjects enrolled in clinical trials of
medicinal products, to ensure quality of conduct, and to
standardize regulation and conduct of clinical trials across
Europe. Clinical trials of medicinal products should adhere
to current legislation, regulation and guidance including the
principles of Good Clinical Practice (GCP); a set of internationally harmonized guidelines to serve the protection of
human rights, safety and dignity whilst also assuring ethical
and scientific integrity. The Directive details both ethical
and scientific quality requirements for all levels of developing a clinical trial for a medicinal product from designing,
conducting, recording and reporting in phase I–IV trials. The
Directive emphasizes that the objective of a clinical trial is
to establish safety and efficacy a therapeutic intervention
using one or more medicinal products. A clinical trial must
obtain favorable opinion from a legally constituted Ethical
Review Board. The conduct of a clinical trial should include
statutory inspections by authorities able to verify levels
of efficacy and safety that are acceptable and compliant
Neuropsychiatric Disease and Treatment 2008:4(5)
with GCP. Within Europe, the pharmacovigilance system
Eudravigilance activates the immediate cessation of a trial
with an unacceptable level of risk, exchanging information
between Member States using shared databases. The possibility of adverse reactions means that sponsors of clinical
trials must be able to provide insurance or indemnity to cover
liability costs. Manufacturing and document archiving are
further legislated in EU DIRECTIVE 2005/28/EC.
EU DIRECTIVE 2004/23/EC on setting
standards of quality and safety
for the donation, procurement,
testing, processing, preservation,
storage, and distribution of human
tissues and cells
The use of human tissues and cells for transplantation has
become a strong focus for therapeutic intervention in PD.
The Directive only applies to the use of human tissue and
cells used for clinical trials to the human body; it does not
apply to in vitro research experiments or use in animal models of disease. The quality and safety of these substances
must be maintained, in particular to prevent transmission of
disease. Donated tissues and cells must be procured, tested,
processed, preserved, and stored in accordance with validated and approved safety measures. In order to regulate the
highest possible quality control and assurance surrounding
the handling, preparation and storage of these substances
for use in clinical trials, laboratory conditions must achieve
Good Manufacturing Practice (GMP) standards. GMP sets
out highly specified conditions of laboratory practice and
regulation put in place to ensure successful production and
quality control of the therapeutic product. In light of the
EU directive, the UK Human Tissue Act (2004) sets out a
new legal framework for the storage and use of tissue from
the living, and the removal, storage and use of tissues and
organs from the dead. The Human Tissue Authority (HTA;
www.hta.gov.uk) is the regulatory body for all matters
concerning the removal, storage, use and disposal of human
tissue for scheduled purposes. The HTA provides advice
and guidance, issues codes of practice and is responsible
for licensing establishments. The fundamental principle
of the HTA is that of consent in relation to the retention
and use of living patients’ organs and tissue for particular
purposes beyond their diagnosis and treatment, and consent
surrounding the removal, retention and use of tissue from
those who have died (where consent is obtained either by
those individuals in life, or after death by someone nominated
841
Lane et al
by or close to them). The practicalities of this legislation are
that GMP standards have to be adhered to in the handling
and processing of human tissue for transplantation. As such,
European groups intending to perform transplantations have
had to install (at their own cost) facilities and equipment and
provide training to gain GMP approval. There is a vastly
more detailed paper trail that will ensure the tracking of
individual samples from derivation at elective termination to
eventual transplantation, and processes previously validated
have to be amended to ensure that all products involved in
the processing of the tissue are of GMP status.
a minimum of 7 years of treatment and the presence of
intractable problems which indicates patients in the more
advanced stages of disease. All these patients had been
taking L-dopa for several years and therefore had established L-dopa induced dyskinesia. Rodent experiments
have demonstrated an association between the severity of
these behaviors or exposure to L-dopa prior to transplantation with the extent of graft-induced dyskinesia (Lane et al
2006). Earlier stage patients are less likely to have developed intractable L-dopa induced dyskinesia and therefore
this may be a more suitable target group of patients.
Considerations in the development
of treatments for PD
Risks and predicted side effects
Beyond the legislative constraints that arise for all disease
treatment strategies using at viral vector or human tissue
methodologies, there are practical and ethical issues pertaining to the treatment of PD that require consideration.
Conclusive diagnosis
A major concern with the treatment of motor disorders and
the successful evaluation of future therapies in small scale
clinical trials is that there is no conclusive pre-mortem diagnosis of PD. Lewy bodies remain the defining pathological
hallmark in addition to the presence of nigrostriatal degeneration, the presence of which cannot be confirmed until
post-mortem. Unfortunately the motor symptoms alone are
unable to provide a conclusive diagnosis of PD as there is
a broad spectrum of phenotypes and a distinct overlap with
Parkinson’s plus syndromes, even responsiveness to L-dopa,
typical of PD, can be confounding in the case of multiple
systems atrophy, as early stage patients can present both with
symptoms indistinguishable from PD and a responsiveness
to L-dopa. Furthermore, even with a confident diagnosis of
PD this in itself incorporates a range of disease phenotypes
which may correspond to subgroups of patients for whom
different treatment approaches may be more appropriate or
more effective (Foltynie et al 2002).
When to intervene?
The stage of the disease at which the trials should be carried out is an ongoing debate. Where existing therapies
are capable of alleviating symptoms it is more complex
to involve these patients in clinical trials of highly novel
therapeutic approaches. However, patients in a more
advanced state may not be the optimum patient group to
benefit from the treatment strategy. Inclusion criteria for
trials of transplantation of embryonic cells have included
842
The risks to patients undergoing primary or stem cell therapy
must all be considered and eliminated or minimized in preclinical experimentation prior to initiation of a clinical trial,
however this is not possible. Some patients that received
dopaminergic fetal tissue transplants into the striatum
developed the unpredicted side effect of ‘off ’ medication
dyskinesia. While the reasons for this side effect are becoming clearer it heightens the need for careful observation to
identify unexpected side effects both on and off medication.
This development has hindered the progress of primary and
stem cell transplantations and will also impact upon viral
vector clinical trials. Other risks to patients undergoing cell
transplantation include the risk of tumor formation, the migration of cells to inappropriate brain regions, immune rejection
of the cells in addition to the inherent risks of neurosurgery
such as hemorrhage and post-operative infection.
Gene therapy carries its own associated risks, and
although huge progress has been made in recent years,
improvements in the safety of viral vectors have to be
demonstrated. The risks include insertional mutagenesis
associated with retroviral vectors (implicated in the development of leukemia in X-SCID sufferers in France). This
is avoided with the use of adeno-associated viruses but the
potentially high immunogenicity has also led to recommendations from the UK Gene Therapy Advisory Committee
(GTAC) that more detailed immune status and cytokine
profiling of the patients is carried out throughout the trial
(Relph et al 2004). The first trials carried out with the AAV2
containing neurturin have not demonstrated any adverse
side effects, however this only encompasses a small group
of patients followed over a short period of time. Unlike
cell transplantation, there have been no proof-of-principle
experiments helping guide new clinical trials and long-term
safety and efficacy trials still need to be carried out. The
recent EU directives on good practice in clinical trials and
Neuropsychiatric Disease and Treatment 2008:4(5)
Treatments for PD and current EU legislation
the manufacture of products for clinical use detailed above
have influenced these processes.
Placebo effects and sham surgery
As with other diseases, there is evidence that this patient
group is subject to placebo effects (de la Fuente-Fernandez
and Stoessl 2002). This is commonly seen with non-invasive
drug treatments but has also been demonstrated with DBS
(de la Fuente-Fernandez 2004). The ethics of blinded sham
surgery to demonstrate clinical efficacy is also a contentious
area with differing opinions from scientists, patients and
public health departments (Boer and Widner 2002; Kim et al
2005; Polgar and Ng 2007; Frank et al 2008). Arguments
include the level of risk and associated costs of ‘unnecessary’
surgical procedures against the validity of non-blinded trials
which could lead to false positive results, although the acceptability of this approach to members of the public appears to
depend on their disease status, those without PD tending to
be more willing to risk sham surgery (Frank et al 2008).
Outcome measures
Restorative therapy in PD aims to reinstate brain circuitry
and function lost as a consequence of progressive neurodegenerative disease. The clinically beneficial effect of a
therapy is dependent on two main issues: firstly, whether
the therapy will survive and replace lost neurons in the
place(s) where required, and secondly, whether the therapy
can restore normal function effectively. Assessing efficacy
depends on sensitive and reliable outcome measures, ideally well-validated clinical and biological markers collected
systematically over a longitudinal period. To date, previous
and on-going clinical trials of restorative therapy in PD have
relied on extensive assessment protocols, eg, cell therapies
follow CAPIT: Core Assessment Protocol for Intracerebral
Transplantation, capturing longitudinal data on clinical and
imaging markers. In spite of such existing protocols, there
is a continued demand for the development of more accurate
and reliable outcome measures to assess the scientific and
economic feasibility of future restorative therapy.
There are many new avenues of therapy on the horizon
for the treatment of PD. However, in order for them to successfully reach the clinic as a mainline treatment clinical
trials will have to demonstrate proof of principle, safety and
efficacy, as the primary fetal tissue studies have done. There
is increasing amounts of legislation surrounding the ethical
and practical issues which must be considered in the planning
of new clinical trials as well as adequate designs to ensure
Neuropsychiatric Disease and Treatment 2008:4(5)
conclusive data are obtained and the true potential of these
novel therapies is realized.
Disclosures
None of the authors has any conflicts of interest to
disclose.
References
Baker SA, Baker KA, Hagg T. 2004. Dopaminergic nigrostriatal projections
regulate neural precursor proliferation in the adult mouse subventricular
zone. Eur J Neurosci, 20:575–9.
Bara-Jimenez W, Bibbiani F, Morris MJ, et al. 2005. Effects of serotonin
5-HT1A agonist in advanced Parkinson’s disease. Mov Disord,
20:932–6.
Bara-Jimenez W, Dimitrova T, Sherzai A, et al. 2004. Effect of monoamine
reuptake inhibitor NS 2330 in advanced Parkinson’s disease.
Mov Disord, 19:1183–6.
Ben-Hur T, Idelson M, Khaner H, et al. 2004. Transplantation of human
embryonic stem cell derived neural progenitors improves behavioral
deficit in Parkinsonian rats. Stem Cells, 22:1246–55.
Bernheimer H, Birkmayer W, Hornykiewicz O, et al. 1973. Brain dopamine
and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci, 20:415–55.
Betarbet R, Turner R, Chockkan V, et al. 1997. Dopaminergic neurons
intrinsic to the primate striatum. J Neurosci, 17:6761–8.
Bjorklund A. 1992. Dopaminergic transplants in experimental parkinsonism:
cellular mechanisms of graft-induced functional recovery. Curr Opin
Neurobiol, 2:683–9.
Bjorklund A, Dunnett SB, Brundin P, et al. 2003. Neural transplantation for
the treatment of Parkinson’s disease. Lancet Neurol, 2:437–45.
Boer GJ, Widner H. 2002. Clinical neurotransplantation: core assessment protocol rather than sham surgery as control. Brain Res Bull,
58:547–53.
Brederlau A, Correia AS, Anisimov SV, et al. 2006. Transplantation of
human embryonic stem cell-derived cells to a rat model of Parkinson’s
disease: effect of in vitro differentiation on graft survival and teratoma
formation. Stem Cells, 24:1433–40.
Carlsson T, Bjorklund T, Kirik D. 2007. Restoration of the striatal dopamine synthesis for Parkinson’s disease: viral vector-mediated enzyme
replacement strategy. Curr Gene Ther, 7109–20.
Carlsson T, Winkler C, Burger C, et al. 2005. Reversal of dyskinesias in an
animal model of Parkinson’s disease by continuous L-DOPA delivery
using rAAV vectors. Brain, 128:559–69.
Chaudhuri KR, Healy DG, Schapira AH. 2006. Non-motor symptoms
of Parkinson’s disease: diagnosis and management. Lancet Neurol,
5:235–45.
Choi-Lundberg DL, Lin Q, Schallert T, et al. 1998. Behavioral and cellular
protection of rat dopaminergic neurons by an adenoviral vector encoding
glial cell line-derived neurotrophic factor. Exp Neurol, 154:261–75.
Clarke CE, 2002. Medical management of Parkinson’s disease. J Neurol
Neurosurg Psychiatry, 72(Suppl 1):I22–I27.
Dass B, Olanow C W, Kordower JH. 2006. Gene transfer of trophic factors
and stem cell grafting as treatments for Parkinson’s disease. Neurology,
66:S89–103.
de la Fuente-Fernandez R. 2004. Uncovering the hidden placebo effect in
deep-brain stimulation for Parkinson’s disease. Parkinsonism Relat
Disord, 10:125–7.
de la Fuente-Fernandez R, Stoessl AJ. 2002. The placebo effect in
Parkinson’s disease. Trends Neurosci, 25:302–6.
Dubach M, Schmidt R, Kunkel D, et al. 1987. Primate neostriatal neurons
containing tyrosine hydroxylase: immunohistochemical evidence.
Neurosci Lett, 75:205–10.
Dunnett SB, Bjorklund A, Lindvall O. 2001. Cell therapy in Parkinson’s
disease – stop or go? Nat Rev Neurosci, 2:365–9.
843
Lane et al
Ekesbo A, Andren PE, Gunne LM, et al. 2000. Motor effects of
(-)-OSU6162 in primates with unilateral 6-hydroxydopamine lesions.
Eur J Pharmacol, 389:193–9.
Eslamboli A, Cummings RM, Ridley, et al. 2003. Recombinant adenoassociated viral vector (rAAV) delivery of GDNF provides protection
against 6-OHDA lesion in the common marmoset monkey (Callithrix
jacchus). Exp Neurol, 184:536–48.
Emborg ME, Carbon M, Holden JE, et al. 2007. Subthalamic glutamic acid
decarboxylase gene therapy: changes in motor function and cortical
metabolism. J Cereb Blood Flow Metab, 27:501–9.
Foltynie T, Brayne C, Barker RA. 2002. The heterogeneity of idiopathic
Parkinson’s disease. J Neurol, 249:138–45.
Frackiewicz EJ, Jhee SS, Shiovitz TM, et al. 2002. Brasofensine treatment
for Parkinson’s disease in combination with levodopa/carbidopa.
Ann Pharmacother, 36:225–30.
Frank SA, Wilson R, Holloway RG, et al. 2008. Ethics of sham surgery:
Perspective of patients. Mov Disord, 23:63–8.
Freed CR, Greene PE, Breeze RE, et al. 2001. Transplantation of embryonic
dopamine neurons for severe Parkinson’s disease. N Engl J Med,
344:710–9.
Frielingsdorf H, Schwarz K, Brundin P, Mohapel P, 2004. No evidence for
new dopaminergic neurons in the adult mammalian substantia nigra.
Proc Natl Acad Sci U S A, 101:10177–82.
Fukushima K, Richter WE Jr, Shiota T, 1977. Partial purification of
6-(D-erythro-1’,2’,3’-trihydroxypropyl)-7,8-dihydropterin triphosphate
synthetase from chicken liver. J Biol Chem, 252:5750–5.
Georgievska B, Kirik D, Bjorklund A. 2002. Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine
neurons induced by long-lasting overexpression of glial cell line
derived neurotrophic factor in the striatum by lentiviral gene transfer.
Exp Neurol, 177:461–74.
Gill SS, Patel NK, Hotton GR, et al. 2003. Direct brain infusion of glial
cell line-derived neurotrophic factor in Parkinson disease. Nat Med,
9:589–95.
Goetz CG, Damier P, Hicking C, et al. 2007. Sarizotan as a treatment for
dyskinesias in Parkinson’s disease: a double-blind placebo-controlled
trial. Mov Disord, 22:179–86.
Hagell P, Piccini P, Bjorklund A, et al. 2002. Dyskinesias following neural
transplantation in Parkinson’s disease. Nat Neurosci, 5:627–8.
Herman JP, Abrous ND. 1994. Dopaminergic neural grafts after fifteen
years: results and perspectives. Prog Neurobiol, 44:1–35.
Hoglinger GU, Rizk P, Muriel MP, et al. 2004. Dopamine depletion
impairs precursor cell proliferation in Parkinson disease. Nat Neurosci,
7:726–35.
Jellinger KA, 1991. Pathology of Parkinson’s disease. Changes other than
the nigrostriatal pathway. Mol Chem Neuropathol, 14:153–97.
Kim JH, Auerbach JM, Rodriguez-Gomez JA, et al. 2002. Dopamine
neurons derived from embryonic stem cells function in an animal model
of Parkinson’s disease. Nature, 418:50–6.
Kim SY, Frank S, Holloway R, et al. 2005. Science and ethics of sham
surgery: a survey of Parkinson disease clinical researchers. Arch
Neurol, 62:1357–60.
Kirik D, Rosenblad C, Bjorklund A, Mandel RJ. 2000. 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, 20:4686–700.
Kordower JH, Freeman TB, Snow BJ, et al. 1995. Neuropathological
evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson’s disease.
N Engl J Med, 332:1118–24.
Kordower JH, Palfi S, Chen EY, et al. 1999. Clinicopathological findings
following intraventricular glial-derived neurotrophic factor treatment
in a patient with Parkinson’s disease. Ann Neurol, 46:419–24.
Lane EL, Winkler C, Brundin P, Cenci MA. 2006. The impact of
graft size on the development of dyskinesia following intrastriatal
grafting of embryonic dopamine neurons in the rat. Neurobiol Dis,
22:334–45.
844
Lang AE, Gill S, Patel NK, et al. 2006. Randomized controlled trial of
intraputamenal glial cell line-derived neurotrophic factor infusion in
Parkinson disease. Ann Neurol, 59:459–66.
Lee B, Lee H, Nam YR, et al. 2005. Enhanced expression of glutamate
decarboxylase 65 improves symptoms of rat parkinsonian models.
Gene Ther, 12:1215–22.
Leff SE, Spratt SK, Snyder RO, Mandel RJ. 1999. 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. Neuroscience, 92:185–96.
Li H, He Z, Su T, et al. 2003. Protective action of recombinant neurturin on
dopaminergic neurons in substantia nigra in a rhesus monkey model of
Parkinson’s disease. Neurol Res, 25:263–7.
Lindholm P, Voutilainen MH, Lauren et al. 2007. Novel neurotrophic
factor CDNF protects and rescues midbrain dopamine neurons in vivo.
Nature, 448:73–7.
Lindvall O, Hagell P. 2002. Role of cell therapy in Parkinson disease.
Neurosurg Focus, 13:e2.
Lindvall O, Wahlberg LU. 2008. Encapsulated cell biodelivery of GDNF:
A novel clinical strategy for neuroprotection and neuroregeneration in
Parkinson’s disease? Exp Neurol, 209:82–8.
Luo J, Kaplitt MG, Fitzsimons HL, et al. 2002. Subthalamic GAD gene
therapy in a Parkinson’s disease rat model. Science, 298:425–9.
Ma Y, Feigin A, Dhawan V, et al. 2002. Dyskinesia after fetal cell
transplantation for parkinsonism: a PET study. Ann Neurol,
52:628–34.
MacDonald BK, Cockerell OC, Sander JW, et al. 2000. The incidence and
lifetime prevalence of neurological disorders in a prospective community-based study in the UK. Brain, 123:665–76.
Maeda T, Nagata K, Yoshida Y, et al. 2005. Serotonergic hyperinnervation
into the dopaminergic denervated striatum compensates for dopamine
conversion from exogenously administered l-DOPA. Brain Res,
1046:230–3.
Marks JJ. 2006. Phase 1 study of putaminal gene transfer with adeno-associated virus serotype 2 [AAV2]-neurturin (CERE-120) for Parkinson’s
disease: preliminary observations. American Academy of Neurology
58th Annual Meeting, San Diego, CA, 2006.
Morizane A, Li JY, Brundin P. 2008. From bench to bed: the potential of
stem cells for the treatment of Parkinson’s disease. Cell Tissue Res,
333:323–36.
Muramatsu S, Fujimoto K, Ikeguchi K, et al. 2002. Behavioral recovery
in a primate model of Parkinson’s disease by triple transduction of
striatal cells with adeno-associated viral vectors expressing dopaminesynthesizing enzymes. Hum Gene Ther, 13:345–54.
Nutt JG, Burchiel KJ, Comella CL, et al. 2003. Randomized, double-blind
trial of glial cell line-derived neurotrophic factor (GDNF) in PD.
Neurology, 60:69–73.
Nyholm D, Askmark H, Gomes-Trolin C, et al. 2003. Optimizing levodopa
pharmacokinetics: intestinal infusion versus oral sustained-release
tablets. Clin Neuropharmacol, 26:156–63.
Olanow CW, Goetz CG, Kordower JH, et al. 2003. A double-blind controlled
trial of bilateral fetal nigral transplantation in Parkinson’s disease.
Ann Neurol, 54:403–14.
Park CH, Minn YK, Lee JY, et al. 2005. In vitro and in vivo analyses of
human embryonic stem cell-derived dopamine neurons. J Neurochem,
92:1265–76.
Patel NK, Gill SS. 2007. GDNF delivery for Parkinson’s disease.
Acta Neurochir Suppl, 97:135–54.
Perrier AL, Tabar V, Barberi T, et al. 2004. Derivation of midbrain dopamine
neurons from human embryonic stem cells. Proc Natl Acad Sci U S A,
101:12543–8.
Piccini P, Brooks DJ, Bjorklund A, et al. 1999. Dopamine release from
nigral transplants visualized in vivo in a Parkinson’s patient. Nat
Neurosci, 2:1137–40.
Piccini P, Pavese N, Hagell P, et al. 2005. Factors affecting the clinical
outcome after neural transplantation in Parkinson’s disease. Brain,
128:2977–86.
Neuropsychiatric Disease and Treatment 2008:4(5)
Treatments for PD and current EU legislation
Pirker W, Tedroff J, Ponten H, et al. 2001. Coadministration of (-)-OSU6162
with l-DOPA normalizes preproenkephalin mRNA expression in the
sensorimotor striatum of primates with unilateral 6-OHDA lesions.
Exp Neurol, 169:122–34.
Poewe W, Wenning GK. 2000. Apomorphine: an underutilized therapy for
Parkinson’s disease. Mov Disord, 15:789–94.
Polgar S, Ng J. 2007. A critical analysis of evidence for using sham surgery
in Parkinson’s disease: implications for public health. Aust N Z J Public
Health, 31:270–4.
Porritt MJ, Batchelor PE, Hughes AJ, et al. 2000. New dopaminergic neurons
in Parkinson’s disease striatum. Lancet, 356:44–45.
Redmond DE Jr, Bjugstad KB, Teng YD, et al. 2007. Behavioral improvement in a primate Parkinson’s model is associated with multiple
homeostatic effects of human neural stem cells. Proc Natl Acad Sci
U S A, 104:12175–80.
Relph K, Harrington K, Pandha H. 2004. Recent developments and current
status of gene therapy using viral vectors in the United Kingdom. BMJ,
329:839–42.
Rosenblad C, Georgievska B, Kirik D. 2003. Long-term striatal overexpression of GDNF selectively downregulates tyrosine hydroxylase in the
intact nigrostriatal dopamine system. Eur J Neurosci, 17:260–70.
Rosenblad C, Kirik D, Devaux B, et al. 1999. Protection and regeneration of
nigral dopaminergic neurons by neurturin or GDNF in a partial lesion
model of Parkinson’s disease after administration into the striatum or
the lateral ventricle. Eur J Neurosci, 11:1554–66.
Roy NS, Cleren C, Singh SK, et al. 2006. Functional engraftment of human
ES cell-derived dopaminergic neurons enriched by coculture with
telomerase-immortalized midbrain astrocytes. Nat Med, 12:1259–68.
Sage JI, Mark MH. 1992. The rationale for continuous dopaminergic
stimulation in patients with Parkinson’s disease. Neurology, 42:23–8;
discussion 57–60.
Sajadi A, Bensadoun JC, Schneider BL, et al. 2006. Transient striatal
delivery of GDNF via encapsulated cells leads to sustained behavioral
improvement in a bilateral model of Parkinson disease. Neurobiol
Dis, 22:119–29.
Schapira AH, Bezard E, Brotchie J, et al. 2006. Novel pharmacological targets
for the treatment of Parkinson’s disease. Nat Rev Drug Discov, 5:845–54.
Sherer TB, Fiske BK, Svendsen CN, et al. 2006. Crossroads in GDNF
therapy for Parkinson’s disease. Mov Disord, 21:136–41.
Shingo T, Date I, Yoshida H, Ohmoto T. 2002. Neuroprotective and restorative
effects of intrastriatal grafting of encapsulated GDNF-producing cells in
a rat model of Parkinson’s disease. J Neurosci Res, 69:946–54.
Shoulson I. 1998. DATATOP: a decade of neuroprotective inquiry Parkinson
Study Group. Deprenyl and tocopherol antioxidative therapy of
parkinsonism. Ann Neurol, 44:S160–6.
Neuropsychiatric Disease and Treatment 2008:4(5)
Sun M, Kong L, Wang X. et al. 2004. 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. Hum Gene
Ther, 15:1177–96.
Syed N, Murphy J, Zimmerman T Jr, et al. 1998. Ten years’ experience
with enteral levodopa infusions for motor fluctuations in Parkinson’s
disease. Mov Disord, 13:336–8.
Tanaka H, Kannari K, Maeda T, et al. 1999. Role of serotonergic neurons
in L-DOPA-derived extracellular dopamine in the striatum of
6-OHDA-lesioned rats. Neuroreport, 10:631–4.
Tande D, Hoglinger G, Debeir T, et al. 2006. New striatal dopamine neurons
in MPTP-treated macaques result from a phenotypic shift and not
neurogenesis. Brain, 129:1194–200.
Wang L, Muramatsu S, Lu Y, et al. 2002. Delayed delivery of AAV-GDNF
prevents nigral neurodegeneration and promotes functional recovery in
a rat model of Parkinson’s disease. Gene Ther, 9:381–9.
Wang X, Lu Y, Zhang H, et al. 2004. Distinct efficacy of pre-differentiated
versus intact fetal mesencephalon-derived human neural progenitor cells
in alleviating rat model of Parkinson’s disease. Int J Dev Neurosci,
22:175–83.
Winkler C, Kirik D, Bjorklund A. 2005. Cell transplantation in Parkinson’s
disease: how can we make it work? Trends Neurosci, 28:86–92.
Winkler C, Kirik D, Bjorklund A, Dunnett SB. 2000. Transplantation in
the rat model of Parkinson’s disease: ectopic versus homotopic graft
placement. Prog Brain Res, 127:233–65.
Yang M, Donaldson AE, Marshall CE, et al. 2004. Studies on the differentiation of dopaminergic traits in human neural progenitor cells in vitro
and in vivo. Cell Transplant, 13:535–47.
Yasuhara T, Shingo T, Date I. 2007. Glial cell line-derived neurotrophic
factor (GDNF) therapy for Parkinson’s disease. Acta Med Okayama,
61:51–6.
Yasuhara T, Shingo T, Muraoka K, et al. 2005. Early transplantation of an
encapsulated glial cell line-derived neurotrophic factor producing cell
demonstrating strong neuroprotective effects in a rat model of Parkinson
disease. J Neurosurg, 102:80–9.
Zhao M, Momma S, Delfani K, et al. 2003. Evidence for neurogenesis
in the adult mammalian substantia nigra. Proc Natl Acad Sci U S A,
100:7925–30.
Zietlow R, Lane EL, Dunnett SB, et al. 2008. Human stem cells for CNS
repair. Cell Tissue Res, 331:301–22.
Zurn AD, Widmer HR, Aebischer P. 2001. Sustained delivery of GDNF:
towards a treatment for Parkinson’s disease. Brain Res Brain Res Rev,
36:222–9.
845