You are on page 1of 9

Epilepsia, 50(1):24–32, 2009

doi: 10.1111/j.1528-1167.2008.01743.x

CRITICAL REVIEW AND INVITED COMMENTARY

Gene therapy in epilepsy



Véronique Riban, yHelen L. Fitzsimons, and ∗ Matthew J. During

Neuroscience Program, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio
State University, Biological Research Tower, Columbus, Ohio, U.S.A.; and yNeurologix, Inc., Fort Lee,
New Jersey, U.S.A.

promise for gene therapy of neurological disorders


SUMMARY
due to their neuronal tropism, lack of toxicity, and
Results from animal models suggest gene therapy stable persistence in neurons, which results in
is a promising new approach for the treatment of robust, long-term expression of the transgene.
epilepsy. Several candidate genes such as neuro- rAAV vectors have been recently used in phase I
peptide Y and galanin have been demonstrated in clinical trials of Parkinson’s disease with an excel-
preclinical studies to have a positive effect on sei- lent safety profile.
zure activity. For a successful gene therapy-based Prior to commencement of phase I trials for gene
treatment, efficient delivery of a transgene to tar- therapy of epilepsy, further preclinical studies are
get neurons is also essential. To this end, advances ongoing including evaluation of the therapeutic
have been made in the areas of cell transplantation benefit in chronic models of epileptogenesis, as well
and in the development of recombinant viral as assessment of safety in toxicological studies.
vectors for gene delivery. Recombinant adeno- KEY WORDS: Seizures, Transplantation, Adeno-
associated viral (rAAV) vectors in particular show associated viral vector, Neuropeptide Y, Galanin.

Gene therapy was traditionally defined as an approach to tant seizures despite the development of new antiepileptic
replace the defective copy of a gene with a functional copy drugs. For many of these patients, surgical resection is often
and restore normal function in a cell population. It is an ele- the only effective therapeutic approach available. More-
gant therapeutic approach because it derives directly from over, antiepileptic drugs do not prevent the progression of
our knowledge of the molecular biology of a disease, tar- the disease, and for epileptic patients, seizure management
geting its most upstream level. This approach has proven is often synonymous with lifelong pharmacological treat-
effective in genetic diseases such as hemophilia (Chuah ment, with side effects that can be debilitating, and the risk
et al., 2004), X-linked immunodeficiency (Hacein-Bey- of increasing refractoriness over time.
Abina et al., 2002), and other metabolic disorders. How-
ever, the field of application is indeed broad, including both
simple genetic as well as complex acquired disorders, as
Which Epilepsies Are Good
gene therapy enables either overexpression or knockdown Candidates for Gene Therapy?
(using interfering RNA, antisense, or ribozymes) of genes Approximately 30% of epilepsies are believed to be idio-
within a pathological network and is therefore applicable to pathic or of genetic origin (Berkovic et al., 2006). Most of
any disease for which the cascade of pathophysiological them are complex diseases with both genetic and environ-
events has been identified. There is a significant unmet mental causation, however autosomal dominant mono-
need for new therapeutic approaches in epilepsy. About genic epilepsies have also been identified, with the
one-third of epileptic patients suffer from pharmacoresis- majority resulting from polymorphism in ion channels. A
mutation in the nicotinic acetylcholine receptor a4 was the
Accepted June 6, 2008; Early View publication August 20, 2008.
first autosomal defect identified in epileptic patients with
Address correspondence to Matthew J. During, Neuroscience Pro- nocturnal frontal lobe epilepsy (Steinlein et al., 1995).
gram, Department of Molecular Virology, Immunology and Medical Since then, more than 12 mutations associated with
Genetics, The Ohio State University, Biological Research Tower 9th
floor, Room 940, 460 W. 12th Ave, Columbus, OH, U.S.A. E-mail:
channelopathies have been identified (Berkovic et al.,
m.during@aukland.ac.nz 2006). However, pure monogenic epilepsies are relatively
Wiley Periodicals, Inc. rare, and in complex epilepsies, the impact of environmen-
ª 2008 International League Against Epilepsy tal influences compared to genetic factors is difficult to

24
25
Gene Therapy in Epilepsy

assess. In addition, because of the compensatory mechan- induced after intraparenchymal delivery. The cell popula-
isms that take place in the brain, the link between the muta- tion within the CNS is devoid of antigen-presenting cells
tion and the hyperexcitability phenotype is sometimes with only a very limited lymphatic system present (Hickey,
difficult to identify. This may explain why some genetic 2001). However, an invasive surgery induces the breakage
mouse models reproducing the mutation of a gene some- of the blood brain barrier and the penetration of activated
times fail to develop spontaneous seizures. Although sig- lymphocytes. Therefore, the notion that the brain is immu-
nificant insights into the pathophysiological mechanisms nologically privileged has been somewhat reevaluated
of epilepsy have been gained from these models, the single (Barker & Widner, 2004), and while the immune response
gene mutations often do not reproduce the full cascade of observed in the brain is generally less pronounced than in
events that lead to an epileptic phenotype (Noebels, 1996). other peripheral organs, it remains an important factor in
Finally, it is very difficult to design an approach with gene the choice and design of the technique for gene transfer.
therapy for a disease that often involves a large area of the Other essential factors to be considered are the efficiency
brain simply because of the technical limitations of achiev- of gene delivery, the level and stability of transgene expres-
ing widespread gene transfer. For these reasons, genetic sion, and the ability to regulate transgene expression. Dif-
forms of epilepsy are among the most challenging and may ferent techniques have been used to express a gene in a
not be the most suitable initial targets for development of specific region of the brain: Cells transplantation in an
gene therapy-based treatments. Focal epilepsies, and in ex vivo approach (fetal cells, immortalized cells,
particular temporal lobe epilepsy, appear to be better candi- fibroblasts), nonviral vector delivery, liposomes, and viral
dates for gene therapy. The physiopathology of temporal vector delivery including herpes simplex virus, retrovirus
lobe epilepsy has been well studied in animal models, as and lentivirus, adenovirus, and adeno-associated virus.
well as from the analysis of surgical resection tissue, and
several candidate genes have been identified as potential Gene delivery vehicles
therapeutic targets (Vezzani, 2004). Furthermore, the epi- Lipid-based systems of gene delivery are the simplest
leptogenic area can be well defined by imaging and record- technique for gene delivery. Their main advantages are a
ing techniques. Gene therapy allows specific targeting of high loading capacity, low immunogenicity, and the trans-
the epileptogenic region, thus sparing the surrounding fection of nondividing cells (Ewert et al., 2004; Rettig &
healthy tissue and minimizing side effects that often go Rice, 2007). However, gene expression is inefficient and
hand in hand with antiepileptic drug treatment. transient, and they are yet not suitable for gene therapy in
neurological disorders. Similarly, nonviral delivery of a
nude DNA is not currently feasible due to the low effi-
Delivery of Genes to the Brain ciency of transfection and the high level of immune
Route of administration response.
Delivery of genetic material to the brain is a technical The main gene transfer techniques used in clinical
challenge due to the presence of the blood brain barrier, application are cell transplantation and cellular transduc-
which limits access to the central nervous system (CNS). tion by viral vectors. Cell transplantation approaches cur-
Intranasal administration is a feasible approach, and trans- rently emphasize the use of stem cells, typically
gene expression in neural cells has been achieved with this embryonic stem (ES) cells or adult stem cells. Their main
method of administration. A proof of principle experiment advantage is the high compatibility of the transplant with
utilizing intranasal delivery of the antiapoptotic gene the host. Additionally, ES cells are pluripotent and can dif-
ICP10PK within a growth-compromised herpes simplex ferentiate into either glia or different neuronal phenotypes
virus vector resulted in transduction of hippocampal neu- (Rathjen & Rathjen, 2001) and can be transfected in vitro
rons, however the level of transgene expression was lim- to express a protein of interest. However, the use of human
ited (Laing et al., 2006). Furthermore, this method is not ES cells in the clinic is limited due to ethical debate over
appropriate for therapies in which transduction of only a destruction of the embryo as well as the potential for gen-
limited region of the brain is required, unless vectors are erating tumors (Riess et al., 2007). The development of
developed that target only selected subpopulations of cells porcine fetal tissue as xenograft material has been pro-
(reviewed by Muzyczka & Warrington, 2005). posed to overcome the limitation of stem cells availability.
An invasive approach, such as stereotactic surgery, is a Xenotransplants have been implanted in patients with neu-
more efficient route for delivery of a therapeutic gene to a rodegenerative diseases (Deacon et al., 1997; Fink et al.,
specific area of the brain, and high levels of transgene 2000). Although the grafts successfully developed synap-
expression can be achieved following injection of a viral tic contacts with host cells (Deacon et al., 1997), their use
vector such as adeno-associated virus (Ruitenberg et al., is still limited because they carry an additional risk of
2002). To date, this is by far the most commonly used infection due to animal pathogens, and the probability that
method of gene delivery to the brain. A major advantage of the graft will be rejected is increased compared to allo-
gene transfer to the brain is the limited immune response geneic grafts (Isacson & Breakefield, 1997).
Epilepsia, 50(1):24–32, 2009
doi: 10.1111/j.1528-1167.2008.01743.x
26
V. Riban et al.

Viral vectors are currently the most promising tools to virus (hCMV) and human CMV/b-actin (CAG) also trans-
directly introduce a gene into the brain, in particular duced glia at a lower frequency. On the other hand, the
herpes simplex virus (HSV), lentivirus, and adeno- cellular human glial fibrillary acidic promoter (hGFAP)
associated virus (AAV). Retroviruses are not a suitable and rat neuron-specific enolase promoter (rNSE) were
gene delivery vehicle for transduction of neuronal cells shown to almost exclusively restrict expression to glia or
because they require the cell to undergo mitosis. Further- neurons, respectively (Jakobsson et al., 2003). Lentiviral
more, the use of retrovirus in gene therapy has raised vectors have been used successfully for therapeutic benefit
safety issues due to the possibility of insertional mutagen- in animal models of neurological disorders. Lentiviral
esis (Hacein-Bey-Abina et al., 2003). HSV, AAV, and len- mediated overexpression of nerve growth factor in choli-
tivirus transduce both dividing and nondividing cells, and nergic neurons improved neuron survival following lesion
the use of cell type-specific promoters allows targeted in rats (Blesch et al., 2005), and the introduction of an RNA
gene transfer to selected populations of neurons. Thus interference (RNAi) targeting human SOD1 into the mus-
further research to optimize the efficacy of these gene cle of mice overexpressing mutant human SOD1 resulted
delivery systems is a reasonable approach towards the in increased survival of motor neurons and a substantially
development of gene-based treatments for neurological extended life span (Ralph et al., 2005).
disorders. Particular attention has been drawn to the use of recom-
HSV allows packaging of approximately 20 kb and has binant AAV (rAAV) vectors for delivery of transgenes to
strong neuronal tropism. In addition, this vector has the the brain after observing a general absence of toxicity,
ability to spread through the nervous system, and injection lack of induction of a cellular immune response, and effi-
of HSV has resulted in widespread distribution of gene cient transduction of the brain in animal models
transduction (Berges et al., 2007). The main limitation to (McCown, 2005; Coura Rdos & Nardi, 2007). A large
the use of HSV is cytotoxicity and elicitation of a cellular number of serotypes have now been isolated from humans
immune response (McMenamin et al., 1998). The devel- and nonhuman primates (Gao et al., 2005), some of which
opment of helper virus-free HSV1, in which genes have been cloned and packaged into recombinant vectors
involved in viral replication have been deleted, decreases and found to display differing tropism for various neuro-
the vector neurotoxicity (Krisky et al., 1998). Recently, nal types and brain areas (Burger et al., 2004; Taymans
further development of the vector has included the use of et al., 2007). Methods have also been developed for manu-
the neuron-specific tyrosine hydroxylase promoter, which facture of extremely pure, high titer preparations, thus
effectively restricts the transduction to a subset population many different rAAV serotypes can now be routinely
of cells (Cao et al., 2008). packaged and purified to this level in the research labora-
Lentiviral vectors hold potential for gene therapy due to tory. When injected into the brain at moderate to high
their ability to integrate into the host chromosome and titers, transgene expression spreading several millimeters
transduce most cell types in the brain, which facilitates a can be consistently achieved with some AAV serotypes,
high level of sustained transgene expression (Jakobsson & including AAV1, AAV5, AAV7, and AAV8 (Burger
Lundberg, 2006). They also have a relatively large cloning et al., 2004; Broekman et al., 2006; Taymans et al.,
capacity of around nine kilobases (Zhao & Lever, 2007). 2007). Conversely, precise stereotactic surgery combined
Lentiviruses are derived from primate or nonprimate with the use of a less efficient serotype (such as rAAV2)
immunodeficiency viruses with human immunodeficiency now provide the means for targeted transduction of a focal
virus (HIV)-based vectors having undergone the most area, such as the hilus or CA1 area of the hippocampus.
development so far. However, safety concerns arise from The rAAV serotypes that have been characterized to date
the potential for recombination events to occur that may have primarily neuronal tropism and are therefore not opti-
generate a replication-competent virus, and therefore more mal for gene therapy of disorders requiring transduction of
vigilant safety measures are required compared with other glial cells. However it is highly possible that among the
viral vectors. These include removal of the virulence genes large number of serotypes that have been cloned, some with
from the packaging plasmids and introduction of the genes glial tropism will be discovered. Further development of
involved in capsid assembly on two separate plasmids to the vector is still needed however, including improvement
reduce the chances of recombination (Zufferey et al., of expression cassettes, which have a packaging limit of
1997). In addition, self-inactivating vectors, with part of approximately 4.7 kb, and the further characterization of
the long terminal repeat (LTR) promoter removed, have cell-specific promoters for restriction of expression to par-
been developed to abolish transcriptional activity upon vec- ticular subclasses of neurons. This is of particular impor-
tor integration. Various promoters have been evaluated in tance for gene therapy of epilepsy, due to the laminar
lentiviral cassettes. When pseudotyped to the glycoprotein nature of the hippocampus with many layers of neurons in
of the vesicular stomatitis virus (VSV-G), most promoters close proximity that have different functions with respect
displayed a pronounced tropism for neurons, although to epileptogenesis. Several promoters have been used with
some pan-specific promoters such as human cytomegalo- rAAV to restrict expression to a subclass of neuron, such as
Epilepsia, 50(1):24–32, 2009
doi: 10.1111/j.1528-1167.2008.01743.x
27
Gene Therapy in Epilepsy

melanin-concentrating hormone (MCH) neurons in the of the disease and maintain focalization of the epilep-
hypothalamus (Van den Pol et al., 2004), yet promoters tic zone.
have not yet been isolated that restrict expression to neuro- One of the first logical targets for gene therapy of epi-
nal subclasses in the hippocampus, such as GABAergic lepsy was the GABAergic system, based on the pharmaco-
neurons in the hilus, or principal neurons in the dentate logically validated approach that an increase in GABA
gyrus. This is a particularly difficult challenge for rAAV levels in the epileptogenic area increases the threshold of
vectors, due to promoter activity contained within the neuronal excitability, hence decreasing seizure occur-
inverted terminal repeats (Flotte et al., 1993). rence. Different techniques of in vitro or in vivo transfec-
Immunization with AAV prior to intracerebral injection tion of glutamic acid decarboxylase (GAD; the enzyme
generates circulating antibodies that can, in some circum- that catalyzes the synthesis of GABA) were used to
stances, limit the transduction of AAV vector if the titer of increase GABA levels in the tissue of interest (Table 1).
neutralizing antibodies is sufficiently high (Peden et al., Transplantation of fetal GABAergic neurons into the sub-
2004). Thus, the potential exists in human patients for stantia nigra (SN), a structure involved in the propagation
rAAV to be neutralized by preexisting antibodies. Without of seizures, induced a transient decrease in seizure sever-
postmortem brain analysis, it is difficult to assess the level ity in the kindling model (Loscher et al., 1998). Similarly,
of transgene expression following rAAV-mediated gene transplantation of engineered mouse cortical neurons and
therapy, however in a phase I clinical trial for Parkinson's glia expressing GAD65 into the SN or piriform cortex
disease involving intrasubthalamic injection of rAAV- showed an anticonvulsant effect (Thompson et al., 2000;
GAD, there was no correlation between the presence of Gernert et al., 2002). Viral vector-based approaches have
preexisting neutralizing antibodies and improvement in also been used to express GAD in cultured rat hippocam-
clinical motor scores (Kaplitt et al., 2007). pal neurons (Liu et al., 2005), but this technique has not
yet been applied in vivo. The different techniques used in
these studies were thus able to induce the expression of
Genes Targeted in Epilepsy exogenous GAD in the epileptic tissue and locally
The goal of gene therapy for epilepsy is to obtain increase GABA levels. However, this expression obtained
not only a sustained anticonvulsant effect, but also an with cell transplantation was only transient. In addition,
antiepileptogenic effect that will block the progression the effects observed are the consequence of a global

Table 1. Summary of studies for gene therapy of epilepsy


Gene Vector Model Authors
Adenosine Cells expressing adenosine Kindling Huber et al., 2001
Myoblasts delivering adenosine Kindling Guttinger et al., 2005
CCK Lipofectin Audiogenic rats Zhang et al., 1997
ICP10PK HSV-2 Kainate ip Laing et al., 2006
GAD Cells expressing GAD65 Kindling Gernert et al., 2002
Fetal cells Kainate icv Shetty & Turner, 2000
Immortalized astrocytes expressing GAD67 In vitro Sacchettoni et al., 1998
Immortalized GABAergic cells Kainate ip Castillo et al., 2006
AAV-GAD67 In vitro Robert et al., 1997
Fibroblasts, GAD65, GAD67 In vitro Ruppert et al., 1993
Cells expressing GAD65 Kindling Thompson et al., 2000
AAV-antisense GABA-A alpha1 Stim. of IC Xiao et al., 1997
Galanin AAV-preprogalanin Kainate ih Lin et al., 2003
AAV-FIB-galanin Kainate ip/stim. of IC McCown, 2006
AAV-FIB-galanin/AAV-galanin Stim. of IC Haberman et al., 2003
GDNF Ad-GDNF Kainate ip Yoo et al., 2006
AAV-GDNF Kindling, SSLSE Kanter-Schlifke et al., 2007
Glut1 HSV1 Kainate ih McLaughlin et al., 2000
HSP72 HSV Kainate ip Yenari et al., 1998
Homer1 AAV SSLSE Klugmann et al., 2005
NPY AAV-preproNPY Kainate ip, kindling Richichi et al., 2004
NPY AAV-preproNPY SSLSE Noé et al., 2008
NR1 AAV – NR1 oral vaccine Kainate ip During et al., 2000
AAV-NR1A/AAV tet off Stim. of IC Haberman et al., 2002

Ad, adenovirus; CCK, cholecystokinin; icv, intracerebroventricular; ih, intrahippocampal; ip, intraperitonneal; SSLSE, self-sustain-
ing limbic status epilepticus; stim, stimulation.

Epilepsia, 50(1):24–32, 2009


doi: 10.1111/j.1528-1167.2008.01743.x
28
V. Riban et al.

increase of GABA levels, and the effect of a strategy tar- injected into the rat hippocampus. Kainic acid-induced
geting a specific cell population are more difficult to pre- seizure activity was significantly decreased, confirming
dict. Indeed, the loss of interneurons and consecutive the antiepileptic effect of galanin in vivo (Lin et al.,
feedback inhibition described in epilepsy is restricted to 2003). Interestingly, administration of rAAV-preprogala-
certain population of interneurons. Conversely, some nin resulted in not only long lasting expression of gala-
interneurons are preserved and are believed to underlie nin, but also in the transport of the neuropeptide along
network synchrony (Bertrand & Lacaille, 2001; Stief the axonal arborization. Haberman et al. (2003) also
et al., 2007). Haberman and colleagues (2002) demon- demonstrated the antiseizure properties of galanin in two
strated the importance of the preferential transduction of a rat seizure models. They fused the fibronectin secretory
neuronal population. In their study, the infusion of an sequence (FIB) onto galanin for constitutive secretion,
rAAV vector coding for a N-methyl D-aspartate receptor AAV-FIB-galanin was evaluated in a model of focal sei-
1 (NR1) cDNA fragment in the antisense orientation zure genesis, which involves electrical stimulation of the
showed preferential transduction of either inhibitory inter- rat inferior collicular column (IC). Preinfusion of AAV-
neurons or primary output neurons depending on the pro- FIB-galanin into the IC increased the threshold for
moter used in the vector construct. Transduction of these seizures. Moreover, following infusion into the hippo-
two different systems had dramatically opposite effects on campus, AAV-FIB-galanin also resulted in suppression
focal seizures (Haberman et al., 2002). This study showed of electrographic and behavioral seizures induced by kai-
the importance of the promoter choice, and more impor- nic acid and also had a neuroprotective effect on the sur-
tantly, demonstrated the utility of rAAV vectors in engi- vival of hilar interneurons (Haberman et al., 2003). In a
neering a precise and cell-targeted gene therapy approach subsequent study, the vector was injected after a series of
to transduce a specific cell population. Recently, Raol and daily stimulations reached a predetermined threshold of
colleagues (2006) used a different approach targeting the seizure activity, that is, in an already hyperexcitable sys-
GABA receptor subunits rather than direct modulation of tem, and the sustained anticonvulsant effect observed
GABA levels. They designed an AAV5 construct coding demonstrated that rAAV-galanin has a robust effect on
for the a1 subunit of the GABA receptor under control of hippocampal hyperactivity (McCown, 2006).
the a4 subunit (GABRA4) promoter, which is upregulated In several animal models of epilepsy, seizure-induced
after status epilepticus. Intrahippocampal injection of this increases of NPY messenger RNA (mRNA) and protein
vector 2 weeks prior to induction of status epilepticus pro- have been observed in the dentate gyrus of the hippocam-
tected against recurrent seizures and demonstrated the pus, suggesting a modulatory role of the neuropeptide on
importance of GABA receptor composition in the devel- neuronal activity (Vezzani et al., 1999). This role was con-
opment of epileptic circuits (Raol et al., 2006). firmed by in vitro data showing that application of NPY to
Over the past decade, the roles of two neuropeptides, hippocampal slices reduces glutamatergic synaptic excita-
neuropeptide Y (NPY) (Noe et al., 2006) and galanin tion (Klapstein & Colmers, 1997), as well as in vivo stu-
(McCown, 2006), as well as the neuromodulator adeno- dies that showed a strong anticonvulsant effect of NPY
sine (Boison, 2007), in the modulation of neuronal excit- mediated by the Y2 and Y5 receptors (Sperk & Herzog,
ability have been established. The observation that 1997; Reibel et al., 2000). In addition, NPY knockout
epileptic seizures induce the release of these neuropep- mice develop spontaneous epileptic seizures, confirming
tides led to the hypothesis that they played an important the importance of NPY in controlling neuronal excitability
role in epileptic activity. Experimental studies further con- (Baraban et al., 1997; Lin et al., 2006; Morris et al.,
firmed their anticonvulsant and neuroprotective role, and 2007). In human tissue from temporal lobe resection,
suggested that these neuropeptides and their receptors NPY-mediated neurotransmission is altered by seizures
constitute an endogenous system to control epileptic activ- (Vezzani et al., 1999; Vezzani & Sperk, 2004), and the
ity. These systems thus appear a promising target for the modulatory role of NPY on epileptic activity has also been
development of new therapeutics and in particular for validated on hippocampal slices (Patrylo et al., 1999). The
gene therapy. effect of chronic overexpression of NPY in the hippocam-
Experimental studies showed that galanin is released pus was examined in the kainic acid model in rats. rAAV1/
during epileptic seizures and has an inhibitory effect on 2 (a pseudotyped vector consisting of a 1:1 mixture of
neuronal activity through presynaptic inhibition of gluta- AAV1 and AAV2 capsid proteins)-mediated gene transfer
matergic transmission, as well as a strong neuroprotec- of preproNPY to the hippocampus delayed seizure onset
tive effect (Mazarati & Lu, 2005). Administration of and dramatically decreased the occurrence of epileptic sei-
galanin (Mazarati et al., 2000; Kokaia et al., 2001) or zures (Richichi et al., 2004). In order to more closely
nonpeptide ligands (Saar et al., 2002) also induces a approximate the effect of rAAV-NPY on epileptogenesis,
robust anticonvulsant effect in animal models of limbic the vector was evaluated in a chronic model of sponta-
seizures. In a study by Lin and colleagues (2003), an neous and progressive temporal lobe epilepsy. In this
rAAV constitutively overexpressing preprogalanin was model, spontaneous seizures develop after recurrent elec-
Epilepsia, 50(1):24–32, 2009
doi: 10.1111/j.1528-1167.2008.01743.x
29
Gene Therapy in Epilepsy

tric stimulation of the hippocampus, and the frequency of I to phase III trials are very encouraging and have proven
seizures increases over time. In rats treated with rAAV1/2- that gene therapy does not present an overall increase in
NPY, progression of seizure activity was repressed, and risk factors associated with the technique compared with
moreover, the frequency of seizures was decreased in other surgical approaches. Gene therapy-based treatments
some animals (Noe et al., 2008). Together these results for neurological disorders including Alzheimer disease
show that AAV-mediated overexpression of NPY shows (Tuszynski et al., 2005), late infantile neuronal ceroid
promise for gene therapy of epilepsy. lipofuscinosis (Worgall et al., 2008), Canavan disease
The inhibitory neuromodulator adenosine has also (McPhee et al., 2006), and Parkinson's disease (Kaplitt
raised interest as an endogenous anticonvulsant (Lee et al., 2007; Fiandaca et al., 2008; Marks et al., 2008)
et al., 1984; Dragunow et al., 1985; Boison et al., 2002). have now been tested in human clinical trials with no
Decreased adenosine levels have been observed in differ- serious adverse events that were attributed to the gene
ent models of epileptogenesis and epileptic activity therapy agent. However, many of the clinical trials did not
(Young & Dragunow, 1994; Gouder et al., 2004; Fedele result in positive results with regard to efficacy.
et al., 2005; Rebola et al., 2005). More recently, adeno- In the first ex vivo gene therapy trial for epilepsy, a
sine has also been shown to restrict the site of epileptogen- xenograft of GABA-expressing cells in a patient candidate
esis via activation of A1 receptors (Fedele et al., 2006). for a temporal lobe resection failed to show an antiepileptic
Using a different approach of ex vivo gene therapy based effect (Diacrin Inc., Charlestown, MA, U.S.A.). Several
on transplantation of cells engineered to release the active issues arise from the use of transplant in neurological dis-
modulator, Boison, Huber, and colleagues showed that eases. In addition to the limited availability of ES cells, the
implantation of encapsulated fibroblasts engineered to survival of the graft is very variable between patients
release adenosine could protect from seizures in the kind- (Bjorklund, 2000). Experimental results in animal models
ling model (Huber et al., 2001). The antiepileptic effect also tend to show a limited survival time of cell transplants
from released adenosine was however transient due to the in the epileptic brain. The temporal lobe is a highly hetero-
short-term survival of the encapsulated fibroblasts. To geneous region organized into complex layers, and the
increase the survival time of the transplant, a recent study type of synaptic connections the graft would develop in
was designed with mouse C2C12 myoblasts genetically this multisynaptic circuit is unknown. In addition, the
engineered to release adenosine by genetic inactivation transplanted cells would be subject to recurrent hyperex-
of adenosine kinase (Guttinger et al., 2005). Intra- citability in the epileptogenic area, which may affect their
ventricular graft of the myoblasts induced a short-term survival. Data obtained from in vivo experiments using
antiepileptic effect on kindling seizures and significantly AAV vectors demonstrate that this method of gene deliv-
reduced seizures duration for a period of 3 weeks after ery may be a more feasible approach for clinical trials. An
transplantation. early proof of principle study demonstrated that gene
Neurotrophic factors play an important role in epilepto- transfer using adeno-associated vector on human resection
genesis (Simonato et al., 2006). Whereas neurogenesis is slices resulted in an appreciable level of cell transduction
increased after status epilepticus and might contribute to of epileptic tissue (O'Connor et al., 1997).
the formation of aberrant circuits, a decrease is observed The therapeutic approach in epilepsy targets a disrup-
during the chronic phase. Glial cell line-derived neuro- tion of the abnormal epileptic activity rather than reintro-
trophic factor (GDNF) administration has been proposed ducing a cell population that has been lost as has often
as a neuroprotective and anticonvulsant approach. To been the focus for gene therapy of neurodegenerative dis-
examine the role of GDNF as a potential target for gene eases. The potentiation of an endogenous system of sei-
therapy, rAAV-GDNF was injected in the hippocampus zure modulation may induce fewer compensatory effects
either before or after status epilepticus, which resulted in a and be more efficient than trying to compensate for a loss
decrease in the severity and the number of seizures of a specific neuronal population. In view of the experi-
(Kanter-Schlifke et al., 2007). Similarly, hippocampal mental data on animal models, the modulation of the endo-
fetal cell pretreated and grafted with fibroblast growth genous system constituted by galanin, NPY, or adenosine
factor-2 (FGF2; in addition with a caspase inhibitor) and appears to be the most likely to translate to clinical trial,
transplanted in the hippocampus of chronically epileptic and indeed following positive results in preclinical stu-
rats also decreased the number of recurrent seizures (Rao dies, a proposal for the treatment of temporal lobe epilepsy
et al., 2007). with rAAV-NPY was presented to the Recombinant DNA
Advisory Committee of the U.S.A. with favorable review
(http://www4.od.nih.gov/oba/RAC/meetings/Sept2004/RA
Toward the Clinic Cagenda092304.pdf).
Currently, more than a thousand clinical trials using In conclusion, the experimental and clinical data
gene therapy have been designed, among which 17 target obtained from other neurological diseases show the feasi-
neurological diseases. The clinical outcomes of the phase bility of gene therapy for epilepsy. However the field of
Epilepsia, 50(1):24–32, 2009
doi: 10.1111/j.1528-1167.2008.01743.x
30
V. Riban et al.

gene therapy is new, and the potential for adverse effects Acknowledgments
is relatively unknown. As with antiepileptic drugs, there is
a possibility of alteration in limbic system function Research was supported by the National Institutes of Health (NIH).
including memory or mood disturbances. Subjects who Conflict of interest: The authors have read the Journal’s guidelines on
are good candidates for temporal lobectomy are an ideal ethical publication, and this review is consistent with those guidelines.
M.J.D. is a founder and consultant for Neurologix, Inc., a public company
population, since the gene transfer would occur in the interested in commercializing neurological gene therapy. V.R. has no
brain region that has been planned for resection, providing conflicts. H.L.F. is a paid employee of Neurologix, Inc.
a built in rescue procedure if the gene therapy was ineffec-
tive or associated with significant adverse events. An
advantage of gene therapy over current drug regimens is
References
the long lasting expression of the therapeutic gene, as well Baraban SC, Hollopeter G, Erickson JC, Schwartzkroin PA, Palmiter
as the ability to target it to only the regions of the brain that RD. (1997) Knock-out mice reveal a critical antiepileptic role for
neuropeptide Y. J Neurosci 17:8927–8936.
it is intended. However the persistence of vector-mediated Barker RA, Widner H. (2004) Immune problems in central nervous
transgene expression is a double-edged sword; if expres- system cell therapy. NeuroRx 1:472–481.
sion escapes from the targeted area into another brain area, Berges BK, Wolfe JH, Fraser NW. (2007) Transduction of brain by
herpes simplex virus vectors. Mol Ther 15:20–29.
there is a chance of unanticipated negative effects that Berkovic SF, Mulley JC, Scheffer IE, Petrou S. (2006) Human epilepsies:
may not be easily remedied. For this reason, for a gene interaction of genetic and acquired factors. Trends Neurosci 29:391–
therapy product to reach phase I clinical trials, it must pass 397.
Bertrand S, Lacaille JC. (2001) Unitary synaptic currents between
through rigorous animal testing for safety and efficacy at lacunosum-moleculare interneurones and pyramidal cells in rat
different dose levels, including but not limited to compre- hippocampus. J Physiol 532(Pt 2):369–384.
hensive assessments of general health, behavior, organ Bjorklund A. (2000) Cell replacement strategies for neurodegenerative
disorders. Novartis Found Symp 231:7–15.
histology, and vector biodistribution. Blesch A, Conner J, Pfeifer A, Gasmi M, Ramirez A, Britton W, Alfa R,
Importantly, results of human clinical trials of neuro- Verma I, Tuszynski MH. (2005) Regulated lentiviral NGF gene trans-
logical disorders have been very promising with excellent fer controls rescue of medial septal cholinergic neurons. Mol Ther
11:916–925.
safety profiles (Fiandaca et al., 2008). In the first gene Boison D, Huber A, Padrun V, Deglon N, Aebischer P, Mohler H. (2002)
therapy trial for a neurodegenerative disorder, AAV- Seizure suppression by adenosine-releasing cells is independent of
aspartoacylase was administered intraparenchymally to seizure frequency. Epilepsia 43:788–796.
Boison D. (2007) Adenosine-based cell therapy approaches for pharma-
10 children with Canavan disease and was well-tolerated coresistant epilepsies. Neurodegener Dis 4:28–33.
with minimal inflammatory or immune response (McPhee Broekman ML, Comer LA, Hyman BT, Sena-Esteves M. (2006) Adeno-
et al., 2006). Moreover, in two recently completed clinical associated virus vectors serotyped with AAV8 capsid are more effi-
cient than AAV-1 or -2 serotypes for widespread gene delivery to the
trials for AAV-mediated gene therapy of Parkinson's dis- neonatal mouse brain. Neuroscience 138:501–510.
ease, there were no adverse events relating to the gene Burger C, Gorbatyuk OS, Velardo MJ, Peden CS, Williams P, Zolotukhin
therapy (Kaplitt et al., 2007; Marks et al., 2008), and S, Reier PJ, Mandel RJ, Muzyczka N. (2004) Recombinant AAV
viral vectors pseudotyped with viral capsids from serotypes 1, 2 and 5
improvements in Parkinsonian symptoms were also display differential efficiency and cell tropism after delivery to differ-
observed. In the first study, unilateral administration of ent regions of the central nervous system. Mol Ther 10:302–317.
AAV2-GAD to the subthalamic nucleus of 12 Parkinson's Cao HY, Zhang GR, Wang X, Kong L, Geller AI. (2008) Enhanced nigros-
triatal neuron-specific, long-term expression by using neural-specific
patients resulted in a significant improvement in clinical promoters in combination with targeted gene transfer by modified
motor scores up to at least 12 months after surgery (Kaplitt helper virus-free HSV-1 vector particles. BMC Neurosci 9:37.
et al., 2007). Fluorodeoxyglucose positron emission Castillo CG, Mendoza S, Freed WJ, Giordano M. (2006) Intranigral
transplants of immortalized GABAergic cells decrease the expression
photometry also revealed reductions in thalamic motor of kainic acid-induced seizures in the rat. Behav Brain Res 171:109–
cortex activity on the injected side of the brain, which cor- 115.
related with clinical rating scores (Feigin et al., 2007). Chuah MK, Collen D, Vandendriessche T. (2004) Preclinical and clinical
gene therapy for haemophilia. Haemophilia 10(Suppl.4):119–125.
Similarly, following bilateral administration of AAV2- Coura Rdos S, Nardi NB. (2007) The state of the art of adeno-associated
neurturin to the putamen of 12 Parkinson's patients, motor virus-based vectors in gene therapy. Virol J 16:99.
function was also improved at 1 year following surgery Deacon T, Schumacher J, Dinsmore J, Thomas C, Palmer P, Kott S,
Edge A, Penney D, Kassissieh S, Dempsey P and others. (1997)
(Marks et al., 2008). Randomized, controlled phase II Histological evidence of fetal pig neural cell survival after transplan-
trials are now underway for both treatments. tation into a patient with Parkinson's disease. Nat Med 3:350–353.
Taken together, the relative low risk associated with Dragunow M, Goddard GV, Laverty R. (1985) Is adenosine an endogen-
ous anticonvulsant? Epilepsia 26:480–437.
gene therapy and the promising preclinical data on both During MJ, Symes CW, Lawlor PA, Lin J, Dunning J, Fitzsimons HL,
NPY and galanin gene transfer in experimental animal Poulsen D, Leone P, Xu R, Dicker BL and others. (2000) An oral vac-
models suggest that temporal lobe epilepsy, a disease cine against NMDAR1 with efficacy in experimental stroke and epi-
lepsy. Science 287:1453–1460.
clearly refractory to a traditional pharmacological Ewert K, Slack NL, Ahmad A, Evans HM, Lin AJ, Samuel CE, Safinya
approach, is an ideal candidate with gene therapy likely to CR. (2004) Cationic lipid-DNA complexes for gene therapy: under-
have a significant impact on disease management within standing the relationship between complex structure and gene deliv-
ery pathways at the molecular level. Curr Med Chem 11:133–149.
the coming decade.
Epilepsia, 50(1):24–32, 2009
doi: 10.1111/j.1528-1167.2008.01743.x
31
Gene Therapy in Epilepsy

Fedele DE, Gouder N, Guttinger M, Gabernet L, Scheurer L, Rulicke T, Klugmann M, Symes CW, Leichtlein CB, Klaussner BK, Dunning J,
Crestani F, Boison D. (2005) Astrogliosis in epilepsy leads to overex- Fong D, Young D, During MJ. (2005) AAV-mediated hippocampal
pression of adenosine kinase, resulting in seizure aggravation. Brain expression of short and long Homer 1 proteins differentially affect
128(Pt 10):2383–2395. cognition and seizure activity in adult rats. Mol Cell Neurosci
Fedele DE, Li T, Lan JQ, Fredholm BB, Boison D. (2006) Adenosine A1 28:347–360.
receptors are crucial in keeping an epileptic focus localized. Exp Neu- Kokaia M, Holmberg K, Nanobashvili A, Xu ZQ, Kokaia Z, Lendahl U,
rol 200:184–190. Hilke S, Theodorsson E, Kahl U, Bartfai T, Lindvall O, Hçkfelt T.
Feigin A, Kaplitt MG, Tang C, Lin T, Mattis P, Dhawan V, During MJ, (2001) Suppressed kindling epileptogenesis in mice with ectopic
Eidelberg D. (2007) Modulation of metabolic brain networks after overexpression of galanin. Proc Natl Acad Sci USA 98:14006–14011.
subthalamic gene therapy for Parkinson's disease. Proc Natl Acad Sci Krisky DM, Wolfe D, Goins WF, Marconi PC, Ramakrishnan R, Mata
USA 104:19559–19564. M, Rouse RJ, Fink DJ, Glorioso JC. (1998) Deletion of multiple
Fiandaca M, Forsayeth J, Bankiewicz K. (2008) Current status of gene immediate-early genes from herpes simplex virus reduces cytotoxi-
therapy trials for Parkinson's disease. Exp Neurol 209:51–57. city and permits long-term gene expression in neurons. Gene Ther
Fink JS, Schumacher JM, Ellias SL, Palmer EP, Saint-Hilaire M, 5:1593–1603.
Shannon K, Penn R, Starr P, VanHorne C, Kott HS and others. (2000) Laing JM, Gober MD, Golembewski EK, Thompson SM, Gyure KA,
Porcine xenografts in Parkinson's disease and Huntington's disease Yarowsky PJ, Aurelian L. (2006) Intranasal administration of the
patients: preliminary results. Cell Transplant 9:273–278. growth-compromised HSV-2 vector DeltaRR prevents kainate-
Flotte TR, Afione SA, Solow R, Drumm ML, Markakis D, Guggino WB, induced seizures and neuronal loss in rats and mice. Mol Ther
Zeitlin PL, Carter BJ. (1993) Expression of the cystic fibrosis trans- 13:870–881.
membrane conductance regulator from a novel adeno-associated Lee KS, Schubert P, Heinemann U. (1984) The anticonvulsive action of
virus promoter. J Biol Chem 268:3781–3790. adenosine: a postsynaptic, dendritic action by a possible endogenous
Gao G, Vandenberghe LH, Wilson JM. (2005) New recombinant sero- anticonvulsant. Brain Res 321:160–164.
types of AAV vectors. Curr Gene Ther 5:285–297. Lin EJ, Richichi C, Young D, Baer K, Vezzani A, During MJ. (2003)
Gernert M, Thompson KW, Loscher W, Tobin AJ. (2002) Genetically Recombinant AAV-mediated expression of galanin in rat hippocam-
engineered GABA-producing cells demonstrate anticonvulsant pus suppresses seizure development. Eur J Neurosci 18:2087–2092.
effects and long-term transgene expression when transplanted into Lin EJ, Young D, Baer K, Herzog H, During MJ. (2006) Differential
the central piriform cortex of rats. Exp Neurol 176:183–192. actions of NPY on seizure modulation via Y1 and Y2 receptors: evi-
Gouder N, Scheurer L, Fritschy JM, Boison D. (2004) Overexpression of dence from receptor knockout mice. Epilepsia 47:773–780.
adenosine kinase in epileptic hippocampus contributes to epilepto- Liu W, He X, Cao Z, Sheng J, Liu H, Li Z, Li W. (2005) Efficient thera-
genesis. J Neurosci 24:692–701. peutic gene expression in cultured rat hippocampal neurons mediated
Guttinger M, Padrun V, Pralong WF, Boison D. (2005) Seizure suppres- by human foamy virus vectors: a potential for the treatment of neuro-
sion and lack of adenosine A1 receptor desensitization after focal logical diseases. Intervirology 48:329–335.
long-term delivery of adenosine by encapsulated myoblasts. Exp Loscher W, Ebert U, Lehmann H, Rosenthal C, Nikkhah G. (1998) Sei-
Neurol 193:53–64. zure suppression in kindling epilepsy by grafts of fetal GABAergic
Haberman R, Criswell H, Snowdy S, Ming Z, Breese G, Samulski R, neurons in rat substantia nigra. J Neurosci Res 51:196–209.
McCown T. (2002) Therapeutic liabilities of in vivo viral vector trop- Lowenstein PR, Castro MG. (2003) Inflammation and adaptive immune
ism: adeno-associated virus vectors, NMDAR1 antisense, and focal responses to adenoviral vectors injected into the brain: peculiarities,
seizure sensitivity. Mol Ther 6:495–500. mechanisms, and consequences. Gene Ther 10:946–954.
Haberman RP, Samulski RJ, McCown TJ. (2003) Attenuation of seizures Marks WJ Jr, Ostrem JL, Verhagen L, Starr PA, Larson PS, Bakay RA,
and neuronal death by adeno-associated virus vector galanin expres- Taylor R, Cahn-Weiner DA, Stoessl AJ, Olanow CW, Bartus RT.
sion and secretion. Nat Med 9:1076–1080. (2008) Safety and tolerability of intraputaminal delivery of CERE-
Hacein-Bey-Abina S, Le Deist F, Carlier F, Bouneaud C, Hue C, De 120 (adeno-associated virus serotype 2-neurturin) to patients with
Villartay JP, Thrasher AJ, Wulffraat N, Sorensen R, Dupuis-Girod S, idiopathic Parkinson's disease: an open-label, phase I trial. Lancet
Fischer A, Davies EG, Kuis W, Leiva L, Cavazzana-Calvo M. (2002) Neurol 7:400–408.
Gene therapy of X-linked severe combined immunodeficiency. N Mazarati AM, Hohmann JG, Bacon A, Liu H, Sankar R, Steiner RA,
Engl J Med 346:1185–1193. Wynick D, Wasterlain CG. (2000) Modulation of hippocampal excit-
Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, ability and seizures by galanin. J Neurosci 20:6276–6281.
Wulffraat N, Leboulch P, Lim A, Osborne CS, Pawliuk R, Morillon E Mazarati A, Lu X. (2005) Regulation of limbic status epilepticus by hip-
and others. (2003) LMO2-associated clonal T cell proliferation in pocampal galanin type 1 and type 2 receptors. Neuropeptides
two patients after gene therapy for SCID-X1. Science 302:415–419. 39:277–280.
Hickey WF. (2001) Basic principles of immunological surveillance of McCown TJ. (2005) Adeno-associated virus (AAV) vectors in the CNS.
the normal central nervous system. Glia 36:118–124. Curr Gene Ther 5:333–338.
Huber A, Padrun V, Deglon N, Aebischer P, Mohler H, Boison D. (2001) McCown TJ. (2006) Adeno-associated virus-mediated expression and
Grafts of adenosine-releasing cells suppress seizures in kindling epi- constitutive secretion of galanin suppresses limbic seizure activity in
lepsy. Proc Natl Acad Sci USA 98:7611–7616. vivo. Mol Ther 14:63–68.
Isacson O, Breakefield XO. (1997) Benefits and risks of hosting animal McLaughlin J, Roozendaal B, Dumas T, Gupta A, Ajilore O, Hsieh J,
cells in the human brain. Nat Med 3:964–969. Ho D, Lawrence M, McGaugh JL, Sapolsky R. (2000) Sparing of
Jakobsson J, Ericson C, Jansson M, Bjork E, Lundberg C. (2003) neuronal function postseizure with gene therapy. Proc Natl Acad Sci
Targeted transgene expression in rat brain using lentiviral vectors. U S A 97:12804–12809.
J Neurosci Res 73:876–885. McMenamin MM, Byrnes AP, Charlton HM, Coffin RS, Latchman DS,
Jakobsson J, Lundberg C. (2006) Lentiviral vectors for use in the central Wood MJA. (1998) A gamma34.5 mutant of herpes simplex 1 causes
nervous system. Mol Ther 13:484–493. severe inflammation in the brain. Neuroscience 83:1225–1237.
Kanter-Schlifke I, Georgievska B, Kirik D, Kokaia M. (2007) Seizure McPhee SW, Janson CG, Li C, Samulski RJ, Camp AS, Francis J, Shera
suppression by GDNF gene therapy in animal models of epilepsy. D, Lioutermann L, Feely M, Freese A and others. (2006) Immune
Mol Ther 15:1106–1113. responses to AAV in a phase I study for Canavan disease. J Gene
Kaplitt MG, Feigin A, Tang C, Fitzsimons HL, Mattis P, Lawlor PA, Med 8:577–588.
Bland RJ, Young D, Strybing K, Eidelberg D and others. (2007) Morris MJ, Gannan E, Stroud LM, Beck-Sickinger AG, O'Brien TJ.
Safety and tolerability of gene therapy with an adeno-associated virus (2007) Neuropeptide Y suppresses absence seizures in a genetic rat
(AAV) borne GAD gene for Parkinson's disease: an open label, phase model primarily through effects on Y receptors. Eur J Neurosci
I trial. Lancet 369:2097–2105. 25:1136–1143.
Klapstein GJ, Colmers WF. (1997) Neuropeptide Y suppresses epilepti- Muzyczka N, Warrington KH Jr. (2005) Custom adeno-associated virus
form activity in rat hippocampus in vitro. J Neurophysiol 78:1651– capsids: the next generation of recombinant vectors with novel trop-
1661. ism. Hum Gene Ther 16:408–416.

Epilepsia, 50(1):24–32, 2009


doi: 10.1111/j.1528-1167.2008.01743.x
32
V. Riban et al.

Noe F, Nissinen J, Pitkanen A, Gobbi M, Sperk G, During M, Vezzani A. astrocytes transduced by a glutamic acid decarboxylase-expressing
(2006) Gene therapy in epilepsy: the focus on NPY. Peptides 28:377– retrovirus. Glia 22:86–93.
383. Shetty AK, Turner DA. (2000) Fetal hippocampal grafts containing CA3
No F, Pool AH, Nissinen J, Gobbi M, Bland R, Rizzi M, Balducci C, cells restore host hippocampal glutamate decarboxylase-positive
Ferraguti F, Sperk G, During MJ, Pitknen A, Vezzani A. (2008) interneuron numbers in a rat model of temporal lobe epilepsy. J Neu-
Neuropeptide Y gene therapy decreases chronic spontaneous seizures rosci 20:8788–8801.
in a rat model of temporal lobe epilepsy. Brain 131:1506–1515. Simonato M, Tongiorgi E, Kokaia M. (2006) Angels and demons:
Noebels JL. (1996) Targeting epilepsy genes. Neuron 16:241–244. neurotrophic factors and epilepsy. Trends Pharmacol Sci 27:631–
O’Connor WM, Davidson BL, Kaplitt MG, Abbey MV, During MJ, 638.
Leone P, Langer D, O'Connor MJ, Freese A. (1997) Adenovirus vec- Sperk G, Herzog H. (1997) Anticonvulsant action of neuropeptide Y.
tor-mediated gene transfer into human epileptogenic brain slices: pro- Neuropeptide Y may act as an endogenous anticonvulsant through
spects for gene therapy in epilepsy. Exp Neurol 148:167–178. Y5 receptors suggesting a new target for antiepileptic drugs. Nat Med
Patrylo PR, Van Den Pol AN, Spencer DD, Williamson A. (1999) NPY 3:728–729.
inhibits glutamatergic excitation in the epileptic human dentate Steinlein OK, Mulley JC, Propping P, Wallace RH, Phillips HA,
gyrus. J Neurophysiol 82:478–483. Sutherland GR, Scheffer IE, Berkovic SF. (1995) A missense muta-
Peden CS, Burger C, Muzyczka N, Mandel RJ. (2004) Circulating anti- tion in the neuronal nicotinic acetylcholine receptor alpha 4 subunit is
wild-type adeno-associated virus type 2 (AAV2) antibodies inhibit associated with autosomal dominant nocturnal frontal lobe epilepsy.
recombinant AAV2 (rAAV2)-mediated, but not rAAV5-mediated, Nat Genet 11:201–203.
gene transfer in the brain. J virol 78:6344–6359. Stief F, Zuschratter W, Hartmann K, Schmitz D, Draguhn A. (2007)
Ralph GS, Radcliffe PA, Day DM, Carthy JM, Leroux MA, Lee DC, Enhanced synaptic excitation-inhibition ratio in hippocampal inter-
Wong LF, Bilsland LG, Greensmith L, Kingsman SM, Mitrophanous neurons of rats with temporal lobe epilepsy. Eur J Neurosci 25:519–
KA, Mazarakis ND, Azzouz M. (2005) Silencing mutant SOD1 using 528.
RNAi protects against neurodegeneration and extends survival in an Taymans JM, Vandenberghe LH, Haute CV, Thiry I, Deroose CM,
ALS model. Nat Med 11:429–433. Mortelmans L, Wilson JM, Debyser Z, Baekelandt V. (2007) Com-
Rao MS, Hattiangady B, Rai KS, Shetty AK. (2007) Strategies for pro- parative analysis of adeno-associated viral vector serotypes 1, 2, 5, 7,
moting anti-seizure effects of hippocampal fetal cells grafted into the and 8 in mouse brain. Hum Gene Ther 18:195–206.
hippocampus of rats exhibiting chronic temporal lobe epilepsy. Neu- Thompson K, Anantharam V, Behrstock S, Bongarzone E, Campagnoni
robiol Dis 27:117–132. A, Tobin AJ. (2000) Conditionally immortalized cell lines, engi-
Raol YH, Lund IV, Bandyopadhyay S, Zhang G, Roberts DS, Wolfe JH, neered to produce and release GABA, modulate the development of
Russek SJ, Brooks-Kayal AR. (2006) Enhancing GABA(A) receptor behavioral seizures. Exp Neurol 161:481–489.
alpha 1 subunit levels in hippocampal dentate gyrus inhibits epilepsy Tuszynski MH, Thal L, Pay M, Salmon DP, U HS, Bakay R, Patel P,
development in an animal model of temporal lobe epilepsy. J Neu- Blesch A, Vahlsing HL, Ho G and others. (2005) A phase 1 clinical
rosci 26:11342–11346. trial of nerve growth factor gene therapy for Alzheimer disease. Nat
Rathjen J, Rathjen PD. (2001) Mouse ES cells: experimental exploitation Med 11:551–555.
of pluripotent differentiation potential. Curr Opin Genet Dev Van Den Pol AN, Acuna-Goycolea C, Clark KR, Ghosh PK. (2004) Phy-
11:587–594. siological properties of hypothalamic MCH neurons identified with
Rebola N, Porciuncula LO, Lopes LV, Oliveira CR, Soares-da-Silva P, selective expression of reporter gene after recombinant virus infec-
Cunha RA. (2005) Long-term effect of convulsive behavior on the tion. Neuron 42:635–652.
density of adenosine A1 and A 2A receptors in the rat cerebral cortex. Vezzani A, Sperk G, Colmers WF. (1999) Neuropeptide Y: emerging
Epilepsia 46(Suppl 5):159–165. evidence for a functional role in seizure modulation. Trends Neurosci
Reibel S, Larmet Y, Carnahan J, Marescaux C, Depaulis A. (2000) Endo- 22:25–30.
genous control of hippocampal epileptogenesis: a molecular cascade Vezzani A. (2004) Gene therapy in epilepsy. Epilepsy Curr 4:87–90.
involving brain-derived neurotrophic factor and neuropeptide Y. Epi- Vezzani A, Sperk G. (2004) Overexpression of NPY and Y2 receptors in
lepsia 41(Suppl 6):S127–133. epileptic brain tissue: an endogenous neuroprotective mechanism in
Rettig GR, Rice KG. (2007) Non-viral gene delivery: from the needle to temporal lobe epilepsy? Neuropeptides 38:245–252.
the nucleus. Expert Opin Biol Ther 7:799–808. Worgall S, Sondhi D, Hackett NR, Kosofsky B, Kekatpure MV, Neyzi N,
Richichi C, Lin EJ, Stefanin D, Colella D, Ravizza T, Grignaschi G, Dyke JP, Ballon D, Heier L, Greenwald BM, Christos P, Mazumdar
Veglianese P, Sperk G, During MJ, Vezzani A. (2004) Anticonvul- M, Souweidane MM, Kaplitt MG, Crystal RG. (2008) Treatment of
sant and antiepileptogenic effects mediated by adeno-associated virus late infantile neuronal ceroid lipofuscinosis by CNS administration of
vector neuropeptide Y expression in the rat hippocampus. J Neurosci a serotype 2 adeno-associated virus expressing CLN2 cDNA. Hum
24:3051–3059. Gene Ther 19:463–474.
Riess P, Molcanyi M, Bentz K, Maegele M, Simanski C, Carlitscheck C, Xiao X, McCown TJ, Li J, Breese GR, Morrow AL, Samulski RJ. (1997)
Schneider A, Hescheler J, Bouillon B, Schfer U, Neugebauer E. Adeno-associated virus (AAV) vector antisense gene transfer in vivo
(2007) Embryonic stem cell transplantation after experimental trau- decreases GABA(A) alpha1 containing receptors and increases infer-
matic brain injury dramatically improves neurological outcome, but ior collicular seizure sensitivity. Brain Res 756:76–83.
may cause tumors. J Neurotrauma 24:216–225. Yenari MA, Fink SL, Sun GH, Chang LK, Patel MK, Kunis DM, Onley
Robert JJ, Bouilleret V, Ridoux V, Valin A, Geoffroy MC, Mallet J, Le D, Ho DY, Sapolsky RM, Steinberg GK. (1998) Gene therapy with
Gal La Salle G. (1997) Adenovirus-mediated transfer of a functional HSP72 is neuroprotective in rat models of stroke and epilepsy. Ann
GAD gene into nerve cells: potential for the treatment of neurological Neurol 44:584–591.
diseases. Gene Ther 4:1237–1245. Yoo YM, Lee CJ, Lee U, Kim YJ. (2006) Neuroprotection of adenoviral-
Ruitenberg MJ, Eggers R, Boer GJ, Verhaagen J. (2002) Adeno-asso- vector-mediated GDNF expression against kainic-acid-induced exci-
ciated viral vectors as agents for gene delivery: application in disor- totoxicity in the rat hippocampus. Exp Neurol 200:407–417.
ders and trauma of the central nervous system. Methods 28:182–194. Young D, Dragunow M. (1994) Status epilepticus may be caused by
Ruppert C, Sandrasagra A, Anton B, Evans C, Schweitzer ES, Tobin AJ. loss of adenosine anticonvulsant mechanisms. Neuroscience 58:245–
(1993) Rat-1 fibroblasts engineered with GAD65 and GAD67 261.
cDNAs in retroviral vectors produce and release GABA. J Neuro- Zhang LX, Li XL, Smith MA, Post RM, Han JS. (1997) Lipofectin-facili-
chem 61:768–771. tated transfer of cholecystokinin gene corrects behavioral abnormal-
Saar K, Mazarati AM, Mahlapuu R, Hallnemo G, Soomets U, Kilk K, ities of rats with audiogenic seizures. Neuroscience 77:15–22.
Hellberg S, Pooga M, Tolf BR, Shi TS, Hokfelt T, Wasterlain C, Zhao J, Lever AM. (2007) Lentivirus-mediated gene expression. Meth-
Bartfai T, Langel U. (2002) Anticonvulsant activity of a nonpeptide ods Mol Biol 366:343–355.
galanin receptor agonist. Proc Natl Acad Sci U S A 99:7136–7141. Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. (1997) Multiply
Sacchettoni SA, Benchaibi M, Sindou M, Belin MF, Jacquemont B. attenuated lentiviral vector achieves efficient gene delivery in vivo.
(1998) Glutamate-modulated production of GABA in immortalized Nat Biotechnol 15:871–875.

Epilepsia, 50(1):24–32, 2009


doi: 10.1111/j.1528-1167.2008.01743.x

You might also like