You are on page 1of 10

CONFERENCE PAPER

Gutless adenovirus: last-generation adenovirus


for gene therapy
R Alba
1
, A Bosch
1
and M Chillon
1,2
1
Gene Therapy Laboratory, Department of Biochemistry and Molecular Biology, Center of Animal Biotechnology and Gene Therapy
(CBATEG), Universitat Auto`noma de Barcelona, Bellaterra, Spain; and
2
Institut Catala` de Recerca i Estudis Avancats (ICREA),
Barcelona, Spain
Last-generation adenovirus vectors, also called helper-depen-
dent or gutless adenovirus, are very attractive for gene therapy
because the associated in vivo immune response is highly
reduced compared to rst- and second-generation adenovirus
vectors, while maintaining high transduction efciency and
tropism. Nowadays, gutless adenovirus is administered in
different organs, such as the liver, muscle or the central
nervous system achieving high-level and long-term transgene
expression in rodents and primates. However, as devoid of all
viral coding regions, gutless vectors require viral proteins
supplied in trans by a helper virus. To remove contamination
by a helper virus from the nal preparation, different systems
based on the excision of the helper-packaging signal have
been generated. Among them, Cre-loxP system is mostly
used, although contamination levels still are 0.11% too high
to be used in clinical trials. Recently developed strategies to
avoid/reduce helper contamination were reviewed.
Gene Therapy (2005) 12, S18S27. doi:10.1038/sj.gt.3302612
Keywords: adenovirus; gutless; helper-dependent vectors; in vivo gene therapy
Introduction
Gene therapy for most genetic diseases requires expres-
sion of the therapeutic protein for the whole life of
the patient. In order to be efcient for the treatment
of genetic disorders, a gene therapy vector has to meet
several conditions: (i) safety, which can be better
achieved with a nonintegrative vector, as it avoids the
risk for insertional mutagenesis; (ii) ability to be easily
and inexpensively produced at a large-scale in the
laboratory; (iii) stability in target cells, which is favored
with low-immunogenic vectors; and (iv) high-capacity
allowing the possibility of introducing full-length DNA
sequences for most genes, long full-length cDNAs,
endogenous promoters or additional regulatory
sequences such as enhancers or insulators, which can
provide a tightly regulated expression of the therapeutic
gene, similar to physiologic conditions. Adenoviruses
and especially gutless adenoviruses seem to accomplish
most of these conditions as they are not integrative, they
have a capacity of 36 kb, they can be easily produced at
high titers in the laboratory and can be delivered to a
large mass of cells, and, contrary to the rst-generation
adenovirus in which the expression of wild-type adeno-
viral genes stimulates the immune system, gutless
adenoviruses show long-term stability in many tissues.
As seen in Table 1, adenovirus is one of the rst elections
in clinical trials, being the most used vector in the last
5 years (19992004) (visit www.wiley.co.uk/genmed/
clinical for more information). Nowadays, adenovirus
vectors are applied to treat cancer, monogenic disorders,
vascular diseases and others complications.
Basically, wild-type adenovirus is associated with
mild diseases like conjunctivitis, pharyngitis and, in a
high percentage, with colds or acute respiratory diseases
but not to tumoral pathways or other viral alterations.
1
In
the early 1950s, adenovirus was used for the rst time
as a preventive vaccine for respiratory diseases. These
studies supposed a great advance since they allowed the
knowledge of aspects related to associated immune
responses and adverse secondary effects.
As gene transfer vector, the adenovirus has a high
transfection efciency, both in quiescent and in dividing
cells, it does not integrate; allows easy capsid modication
in order to retarget its tropism to different tissues; and
high titers (up to 10
13
particles/ml) are routinely obtained.
In addition, it is noteworthy that in the last 10 years, new
scale-up adenovirus production systems have been
developed, facilitating its use in human clinical trials.
Well-characterized human serotypes Ad2 and Ad5
from group C are the classic adenoviruses used as
vectors. In order to produce safe and nonreplicative
vectors, rst-generation adenoviral vectors containing
the whole viral genome with the exception of the E1
region were developed.
2
To propagate rst-generation
adenoviruses, several E1-expressing cell lines have been
generated: 293,
3
911,
4
N52.E6
5
and PER.C6.
6
Although E1-deleted vectors cannot replicate in vivo,
residual expression from adenoviral genes triggers a
cytotoxic T lymphocyte (CTL) immune response towards
infected cells,
7
which nally leads to the elimination of
transduced cells and, therefore, to the lost of therapeutic
gene expression.
Correspondence: Dr M Chillon, Gene Therapy Laboratory, Department of
Biochemistry and Molecular Biology, Center of Animal Biotechnology and
Gene Therapy (CBATEG), Edici H, Universitat Auto`noma de Barcelona,
Bellaterra 08193, Spain
Gene Therapy (2005) 12, S18S27
& 2005 Nature Publishing Group All rights reserved 0969-7128/05 $30.00
www.nature.com/gt
To avoid this problem, second-generation adenoviral
vectors combining deletion of different early regions
(E17E3 and E2/E4) were generated (Figure 1). Deletion
of these regions permits to accommodate up to 14 kb and
hence increase the vector cloning capacity.
8,9
However,
second-generation Ad vectors still do not avoid in vivo-
associated immunogenicity and toxicity due to residual
gene expression from remaining viral genes.
Third-generation vectors, called gutless or gutted
Ad, devoid of all coding viral regions were recently
generated. They are also called helper-dependent adeno-
viruses because of the need of a helper adenovirus that
carries all coding regions, and high-capacity adenoviruses
because they can accommodate up to 36 kb of DNA.
Briey, the gutless adenovirus only keeps the 5
0
and 3
0
inverted terminal repeats (ITRs) and the packaging
signal (C) from the wild-type adenovirus.
Vector capsids package efciently only 75105% of the
whole adenovirus genome.
1014
As therapeutic expres-
sion cassettes usually do not reach up to 36 kb, there is a
need to use stuffer DNA in order to complete the genome
size for encapsidation. Initially, it was believed that
stuffer DNAs unique role was the participation in the
packaging of the vector genome. Thus, the rst DNA
stuffer used was from lambda phage, yeast, bacterial and
non-human DNA.
14
However, rst injections of gutless
vectors carrying lambda DNA elicited CTL immune
response because peptides from stuffer were presented
in the cell membrane, and it became evident that DNA
stuffer played an important role in the stabilization of
viral DNA into the cell.
15,16
The rst choice was then
DNA from mammalian and human introns since they
seemed to favor maintenance of the gutless genome into
the cell for long periods of time.
15
For example, intronic
sequences from the HPRT gene containing matrix
attachment regions (MAR) elements have been used as
stuffer DNA showing increased DNA stability and no
induction of a CTL response. Sequences from other
human loci have also been used with similar or better
results.
17
However, not all intronic sequences are appropriate
DNA stuffer and, thus, proper candidates should avoid
the following sequences (follow next characteristics)
(i) coding regions, (ii) repetitive sequences (like alu
sequences), (iii) hot spots for recombination, (iv) regions
that can interfere with the expression of the transgene,
and (v) toxic or immunogenic regions. On the contrary,
MAR that seem to stabilize the adenovirus genome into
the nucleus and permit long periods of gene expression
are recommended.
17
Production of gutless adenovirus vectors
Since gutless vectors are devoid of all viral genes,
proteins needed for its genome replication, packaging
and capsid formation must be supplied in trans. This
is achieved by coinfection of the gutless with a helper
adenovirus. However, since both helper and gutless
vectors have the same viral capsid, separation must be
addressed before purication. Thus far, strategies have
been based on reducing the packaging efciency of the
helper genome compared to the gutless genome, either
by mutating its packaging signal,
1820
by the different
Ad5 genome
First
generation
Second
generation
Helper
Dependent Ad

0 36000 10000 20000 30000


MLP
E1 E3
E4 E2A E2B
L1 L2 L3 L4 L5
ITR ITR
Transgene
Transgene
Transgene
E1
E1 E2 E3
E3
E4
Figure 1 Map of adenovirus serotype 5 genome and different generations of adenoviral vectors. Early transcripts are represented by E1E4 regions and late
transcripts are represented by L1L5 regions. MLP: major late promoter; C: packaging signal.
Table 1 Vectors used in gene therapy clinical trials
Total number of
protocols (2004)
Protocol number
increase (19992004)
Retrovirus 263 83
Adenovirus 258 172
Lipofection 85 10
Adeno-associated virus 25 21
Other viruses 148 100
Others 183 148
Data from www.wiley.co.uk/genmed/clinical.
Gutless adenovirus
R Alba et al
S19
Gene Therapy
size of its genome
13
(genomes bigger or smaller than
the optimal do not package efciently), or by specic
elimination of its packaging signal during viral produc-
tion.
21
Initial strategies consisted in the use of helper-
dependent adenoviruses carrying a defective packaging
signal.
13
These vectors had extensive regions of deleted
viral genome, but they still have some coding regions.
Cotransfection of a wild-type adenovirus as a helper
with this gutless permitted to propagate both adeno-
viruses. After several passages, the gutless adenovirus
was puried by CsCl
2
density gradient. However, high
levels of helper virus and multiple recombinations
between both vectors were detected. This, together with
important complications in large-scale production and
purication, led to the development of new strategies.
An important advance was the specic removal of the
packaging signal by Cre recombinase. Parks and colla-
borators
21
developed a production system where the
helper adenovirus had a packaging signal anked by two
loxP sites, and amplication was performed in Cre
recombinase-expressing cell lines (293Cre). When the
helper adenovirus entered the cell, its packaging signal
was excised preventing the inclusion of its genome into
the viral particle, but retaining all coding regions for the
viral proteins needed to produce the gutless vectors
(Figure 2). Using this system, average helper contamina-
tion ranged over 0.110% compared to gutless vectors.
These levels of helper contamination seem to be due
to the limited efciency of packaging signal excision
associated to low recombinase activity or with low
endogenous levels of recombinases. This is caused either
by adenovirus-mediated host cell shut off or by
cytotoxicity of high levels of Cre recombinase.
22,23
Although the nal level of helper contamination is low,
further reduction is desirable to minimize any potential
toxicity associated with the helper virus, especially when
high doses are required.
24
Other recombinases such as FLP have also been used
to remove the packaging signal. FLP is a yeast site-
specic recombinase, which catalyzes recombination
between frt sites.
25
Philip Ng and collaborators
26
created
new FLP-expressing cell lines (293FLP and 293CreFLP)
and a helper adenovirus with a packaging signal anked
by frt sites. Cre and FLP seem to have equivalent
efciency in the production of gutless adenovirus with a
nal helper contamination of 0.11%. A recent and
elegant improvement in the Cre-loxP system has been
the reversion of the packaging signal of the helper virus
in order to avoid generation of a replication competent
adenovirus (RCA) by recombination with the gutless
vector, which has reduced the helper contamination to
levels down to 0.020.1%.
18
However, Cre and FLP are
bidirectional recombinases that permit excision of the
packaging signal and also its re-entrance, favoring
contamination by the helper adenovirus. Improving
HELPER Ad GUTLESS Ad GENOME
CRE/293 CELL LINE
COTRANSFECTION
HUMAN STUFFER DNA

+
RECOMBINASE
CRE
loxP
Sequences

ADENOVIRUS GENOME
ITR ITR

+
VIRAL
PROTEINS
O.1-10%
HELPER ADENOVIRUS
CONTAMINATION
GUTLESS ADENOVIRUS
ITR
ITR
PLASMID
OR VIRUS
Figure 2 Generation of gutless adenovirus using the Cre/loxP system. Gutless and helper genomes are cotransfected in permissive 293 Cre-expressing cells,
where both genomes are amplied and viral proteins produced. Then, packaging signal of the helpers genome is excised by Cre recombinase, preventing its
packaging into the viral capsid, while gutless genome is still packageable. Efciency of the excision process allows 9099.9% purity of the gutless vector.
Gutless adenovirus
R Alba et al
S20
Gene Therapy
the excision efciency of Cre and FLP in cell lines
also improves the opposite reaction. Thus, the use of
unidirectional recombinases, such as FC31, is an
attractive alternative to reduce contamination levels with
the helper adenovirus. FC31 is a unidirectional recombi-
nase, which recognizes homologous sequences called
attB/attP and shows an excision efciency similar to that
of Cre in 293 cells.
27
When FC31 excises attB/attP-
anked sequences, it creates new sequences called attR/
attL preventing the opposite reaction. Thus, the helper
adenovirus carrying an attB/attP-anked packaging
signal opens the door to a new gutless production
system (authors unpublished results).
Another approach to reduce helper Ad contamination
is based on size-restricted protein IX-deleted helper. pIX
is a hexon-associated protein essential for packaging full-
length viral genomes. A 293 pIX-expressing cell line
permitting the growth of a full-length pIX-defective
helper was generated.
28
The use of gutless vectors with
genomes smaller than 36 kb gives a selective advantage
in the pIX system. Plaque-forming unit assay showed
a reduction by 1000 times of helper contamination
compared to the classical Cre-loxP system. However,
southern blot analysis revealed helper contamination
levels of 0.2% (similar to Cre-loxP), meaning that the
majority of helper adenoviruses was packaged into the
capsid. Combining both Cre and pIX size-restricted
systems allowed to reduce the number of helper
adenovirus particles, but defective helper virions still
remain in the nal preparation.
To address the helper contamination issue, other
strategies are based on using nonadenovirus vectors as
helpers, like herpes simplex virus-1
29
and baculovirus.
30
However, low production efciency
29
and 2% of RCA
generation
30
make their use not feasible in large-scale
production.
As important as the design of a helper-free gutless
adenovirus system is its production in large or industrial
scale. Large-scale production of gutless adenovirus is
complex and less efcient since it requires successive
time-consuming gutless:helper adenovirus coinfections.
Unfortunately, this process increases the risk of reorga-
nizations in the vector genome by homologous recombi-
nation with viral E1 sequences present in permissive 293
cells, which can nally lead to the generation of RCAs. To
avoid this, several laboratories have developed permis-
sive cells, such as N52.E6
5
and PERC6,
6
that do not
contain viral sequences prone to recombine. However, to
achieve large quantities of high quality helper-free
gutless vectors needed for clinical assays, it is funda-
mental to develop and improve the methodology in the
amplication and purication steps as well as in vector
quality. Thus, development of several cell lines able to
grow in suspension, like human 293S,
31
PER.C6,
32
and
293Cre suspension cell lines
18
facilitates large-scale
amplication in bioreactors, although purication meth-
ods like non-ultracentrifuge-based methods and elimina-
tion of helper contamination still need to be improved.
Gutless adenovirus and immune response
Systemic delivery of rst-generation adenoviral vectors
is known to induce a strong hosts immune response,
resulting in the rapid elimination of vector-transduced
cells and the generation of neutralizing antibodies
against the transgene products and the adenovirus
capsid. Both nonspecic innate and adaptive immune
responses are involved when rst- and second-genera-
tion adenoviral vectors are administered (see Schagen
et al
33
for an extensive review). Thus, the innate immune
response is rapidly developed after virus entry by
induction of inammatory gene expression and further
recruitment of macrophages, neutrophil and natural
killer cells, leading to an 8090% of rst-generation
vector removal from the liver in 24 h.
34
Basically, innate
immunity is triggered by the adenovirus particle, is
Ad-dose dependent and does not require viral gene
expression.
3537
In a second step, adaptive cellular and humoral
immune responses are developed about 47 days after
delivery. At this time, a second peak of cytokine and
chemokine gene expression and inammation occurs
leading to lymphocytic inltrates and to the induction
of adenovirus-specic CTL.
35
Initially, cellular immune
response is activated when antigen-presenting cells
(APCs) uptake adenovirus particles, process the particles
into small oligopeptides and present them through the
major histocompatibility complex (MHC) class-I mole-
cules at the cell surface. Further binding of CD8+ T cells
to the MHC class-I/peptide complex induces formation
of Ad-or transgene-product-specic CTLs. Therefore, the
novo synthesis does not seem to be required to initiate the
process.
33
However, for late inammation, the expression
of viral genes still encoded within Ad vectors plays
a signicant role. In immunocompetent hosts, this
response limits the duration of transgene expression
and results in adenovirus vector clearance within a few
weeks of administration.
7,34
On the other hand, adaptive humoral immune
response is initiated by the binding of adenovirus
particles to the surface immunoglobulin of B cells.
33
After internalization and virus processing, the adeno-
virus-derived epitopes are presented at the surface of the
B cell by MHC-II molecules. Exposure of these cells to
cytokines from activated CD4
+
-Th2 helper cells will
result in differentiated plasma cells secreting antibodies
towards the adenoviral capsid.
38
High titers of antibodies
against capsid proteins, either pre-existing because of
previous exposure to natural virus or generated as a
result of vector administration, may inhibit subsequent
dosing with the same vector.
Different strategies to circumvent innate and adaptive
immune responses have been developed. However, most
of them present secondary complications and/or their
use in human patients is questionable. These strategies
include macrophage depletion,
39,40
use of immunosup-
pressive agents (cyclosporin A, cyclophosphamide,
dexamethasone, FK506, Interleukin-12 and deoxyper-
gualin),
4146
use of antibodies to deplete CTLs,
47,48
blockade of costimulatory interactions between APCs,
T and B cells,
4952
intrathymic administration of adeno-
virus,
53
oral tolerization,
54
use of vectors derived from
non-crossreacting serotypes,
55,56
use of adenoviruses
from other species
57,58
and coating vectors with inert
chemicals like polyethylene glycol (PEG).
5961
Diverse in vivo studies in mice suggested that, in the
absence of an immune response, rst-generation adeno-
viral vector DNA is maintained as a stable episome in the
host cell.
41,62,63
Last-generation helper-dependent or
Gutless adenovirus
R Alba et al
S21
Gene Therapy
gutless adenovirus vectors display reduced long-term
toxicity and prolonged transgene expression compared
to rst-generation vectors after administration to per-
ipheral organs of immunologically na ve animals.
56,6468
Lack of coding viral genes may account for reduced
adaptive cellular immune response after systemic deliv-
ery of gutless vectors. Initially, gutless vectors are
capable of transducing dendritic cells and stimulating
Ad-specic T-cell responses, independent of viral gene
transcription.
69
However, the expression of viral genes is
required for T cells to exert their effector functions in the
liver,
70
which possibly explains the vector persistence
and improved transgene expression following transduc-
tion with gutless vectors compared to results with rst-
generation Ad vectors.
As expected, systemic delivery of gutless vectors still
induces adaptive humoral response against the vector
capsid as for rst-generation Ad vectors. Indeed, the
development of Ad-specic antibodies does not con-
tribute to the elimination of Ad-transduced cells and
therefore does not affect the persistence of transgene
expression. However, Ad-specic antibodies will bind
the readministered Ad vector and thereby prevent cell
entry and promote opsonization by macrophages.
Humoral response can also be developed towards
circulating antigen induced by gutless adenoviral trans-
fer.
70
Vectors that mediate transgene expression in APCs
trigger antibody formation because they increase the
probability of neoantigen presentation by APCs,
71
and,
hence, careful selection of tissue-specic promoters may
signicantly improve adenovirus-associated toxicity
proles and diminish or abolish APC transduction and
transgene expression.
72
In addition, systemic adminis-
tration of gutless vectors in a clinical setting might be
inefcient because of the presence of circulating neu-
tralizing antibodies against the same or crossreactive
serotype as a consequence of a natural infection or as a
result of previous vector administration. A large propor-
tion of the human population has signicant levels of
antibodies against adenoviruses;
73
thus, the capacity to
overcome a pre-existing immunity is a fundamental
problem. Different successful strategies to circumvent
pre-existing immunity have been applied, such as the
use of alternative gutless serotypes
15
and the use of a
non-human gutless adenovirus.
58
Thus, administration
of gutless CAV-2 vectors allows the stable, high-level
expression in neurons throughout the rat central nervous
system (CNS).
20
It is noteworthy that even in the
presence of an active peripheral immunization with an
adenovirus that completely eliminates expression from
rst-generation vectors within 60 days, gutless vectors
are able to maintain a long-term transgene expression
in the brain
74,75
due to reduced inammation both in
intensity and in duration in the brains of the preimmu-
nized animals.
As innate immune responses are dependent on the
viral capsid or particle, innate responses stimulated by
gutless vectors are similar to those stimulated by rst-
generation adenovirus vectors. Thus, dose-dependent
acute inammation was reported by Brunetti-Pierri and
colleagues
76
in non-human primates following the
administration of high-dose gutless vectors. However,
innate response, as these recently reported, may be
reduced by PEG modication, probably due to lower
vector uptake by Kupffer cells in vivo.
77,78
Surprisingly, gutless vectors have also been proved to
be very efcient in vaccination, due to their longer
duration of expression, their lower antiviral reactivity
and their higher levels of transgene protein in dendritic
cells compared to the same amount of rst-generation
Ad vectors.
79
In vivo administration of gutless adenovirus
Gutless adenoviruses have been administered in vivo to
different tissues in rodents, dogs and also to non-human
primates. Owing to the high efciency of adenoviruses in
targeting hepatocytes, most of the toxicity studies were
carried out in the liver, following intravenous adminis-
tration of the vector.
68
Gutless vectors have been shown
to express non-immunogenic transgenes during the
whole life of the mouse, while expression driven
by rst-generation adenovirus lasted at most for 3
months.
8082
Therapeutic genes carried by gutless vectors have
been administered to the liver of mouse models for
different diseases, such as hemophilia A and B,
83,84
obesity,
66
familial hypercholesterolemia,
81,8587
ornithine
transcarbamylase deciency,
88
diabetes
89
and chronic
viral hepatitis.
90,91
Therapeutic levels of most proteins
have been documented for a long-term duration. In some
cases the antibody response against a nonendogenous
transgene led to a signicant decrease in the circulating
levels of the therapeutic protein. However, when the
transgene expression was driven by a liver-specic
promoter, the immune response was lower, efcacy of
the therapeutic protein increased and secondary effects
derived from systemic circulation of the therapeutic
protein were undetectable.
90,92
The encouraging results
obtained in rodents had promoted the preclinical studies
in larger animal models. This is the case for hemophilia
A and B dogs,
72,83
which resulted in minimal acute liver
toxicity and transient expression of the transgene
allowing for partial or complete correction of hemo-
philia. As in rodents, the use of liver-specic promoters
avoided the development of neutralizing antibodies
against the therapeutic protein, although the expression
was lower than with a viral ubiquitous promoter.
72
Long-term expression of gutless-driven transgene
was also documented in baboons for more than 1 year;
however, a decrease in the levels of the protein was
observed over time.
56
Toxicity in non-human primates
was assessed using different doses of gutless adenovirus
and the results obtained showed that administration of
high doses of adenovirus (10
13
viral particles/kg)
induced a strong activation of the innate inammatory
response that caused the death of the animal.
76
In order to circumvent the decrease in transgene
expression overtime, readministration with gutless vec-
tors has been attempted in mice using different adeno-
virus serotypes. While readministration with different
serotypes is possible using rst-generation adeno-
virus,
55,56,9395
the level of transgene expression is lower
after the second administration, probably due to cross-
reacting CTLs between the different serotype proteins.
96
However, as gutless vectors do not contain viral genes,
readministration of a different serotype leads to the same
levels of the therapeutic protein as in na ve animals
without showing liver toxicity.
15,81
Liver toxicity was also
Gutless adenovirus
R Alba et al
S22
Gene Therapy
absent after repeated administration of the same ser-
otype of gutless;
67
however, it resulted in a drop of 30-
to 100-fold in transgene expression compared to na ve
animals.
15
Efciency of regulated promoters was also evaluated
using gutless vectors as they offer the possibility of
incorporating all the genetic components of an inducible
system plus other additional sequences such as insula-
tors or silencers that have been shown to avoid leakiness
of the uninduced promoter.
97,98
When these promoters
were driven by rst-generation adenoviruses, only one
induction was possible; a signicant hepatotoxicity was
developed when the animals were treated with the
inducer, followed by a rapid loss of vector DNA.
99
This
was not the case with gutless vectors, where expression
was reinduced three and four times over a period of 2
months. Similar results were obtained with different
inducible systems such as tetracycline, tamoxifen or
mifepristone.
9799
Stability of gutless vectors was evaluated during
hepatocyte cell cycle. Surprisingly, after two-third partial
hepatectomy, gutless vector genomes were reduced by
50%, while rst-generation Ad vector copies were
reduced by 71%. More importantly, episomal plasmid
DNA-injected mice showed a reduction of 99% in DNA
copy number. The decrease in vector copy number/cell
correlated with transgene expression. Several hypotheses
have been proposed to explain the persistence of
episomal gutless vectors in dividing hepatocytes, like
centromeric function of the Ad genome or nuclear
retention activity of the Ad terminal protein, but
conclusive results have not been obtained to date.
100
Adenoviruses target the liver very efciently; how-
ever, they are less effective in other cell types lacking the
CAR or adenovirus receptor. Retargeting of gutless
vectors has been attempted by the incorporation of
polylysine or RGD in the HI loop of the adenoviral ber
protein, which can be obtained by adding ber-modied
helper viruses in the last amplication step of the gutless
vector production. Polylysine has been shown to
effectively transduce mature muscle cells in vitro and
in vivo,
101
while RGD increased the efciency of
transducing ovarian carcinoma cell lines, primary
vascular smooth muscle cells and primary human
endothelial cells.
102
Duchenne muscular dystrophy (DMD), an X-linked
lethal disorder that affects 1 in 3500 males, is caused by
genetic mutations in the dystrophin gene. The cDNA for
full-length dystrophin is approximately of 14 kb, far
above the size of most gene transfer viral vectors. Thus,
the highcapacity of gutless adenoviruses has opened the
possibility to treat DMD animal models
103
not only with
one full-length dystrophin cDNA but also with two
copies of the therapeutic gene.
104,105
In these studies,
neonate skeletal muscles of mdx mice injected with
gutless adenovirus expressed dystrophin for the dura-
tion of the experiment, up to 1 year. At this point, 52%
of the muscle bers showed transgene expression and
functional correction of muscle contractility, and
improved histopathology was reported, correlating with
the level of dystrophin expression. However, a four-fold
decline in the vector DNA copy number was observed
from 10 days to 1 year after vector injection, and the
treated mice developed a signicant humoral response.
No transduced muscle ber loss was reported, indicating
that the decline in DNA copies could be due to instability
of the vector DNA into the host cell.
104
However, loss of
DNA copy number/cell has no consequences in dystro-
phin expression or muscle function in experiments
performed in mice because dystrophin is stable for 26
weeks. When skeletal muscle bers were transduced
before birth (in utero at embryonic day 16), expression of
the reporter protein was stable at least for 5 months
despite the development of antibodies against the
transgene and the adenoviral capsid.
106
However, in this
case, the authors reported no loss of DNA copy number.
Lower levels of toxicity compared to rst-generation
eneration Ad that resulted in higher survival rates of
animals transduced in utero were also documented.
Mdx muscle goes through cycles of necrosis followed
by regeneration, which could account for the higher
efciency of infection of muscle bers from young and
old mdx mice by adenovirus vectors compared to control
animals as reported by the group of JS Chamberlain.
107
On the contrary, the high turnover of muscle cells in the
mdx mice can decrease the stability of nonintegrative
vectors like adenovirus. In 1-year old mdx mice,
transduction of 2530% of skeletal muscle cross-sectional
area with gutless vectors carrying the dystrophin cDNA
under the regulation of a muscle-specic promoter lead
to 40% correction of their high susceptibility of contrac-
tion-induced injury 1 month after treatment.
107
However,
longer time points were not evaluated, but mild immune
cell inltration was reported using mouse dystrophin
cDNA.
Administration of recombinant protein to the CNS
requires the breakdown of the bloodbrain barrier, a
procedure that should not be used for repetitive read-
mnistrations. Thus, permanent transduction of neurons
offers the possibility of stable treatment of diseases
affecting the CNS. Gene transfer to the CNS has been
explored using different vector systems. Despite the
immunoprotection of the CNS, results with rst-genera-
tion adenovirus show a decrease in the expression of the
transgene 2 months after transduction, correlating with
the disappearance of the adenoviral DNA.
108
Acute loss
of transduced cells and chronic inammation could
account for this decline, mainly at high doses.
109
On the
contrary, gutless adenovirus expression can be detected
in the CNS 1 year after striatal injections.
20,110
Moreover
and contrary to what was reported for other tissues,
preimmunization with the same serotype of adenovirus
does not alter gutless stability in the CNS, and does not
signicantly reduce the initial infection of the CNS by
gutless vectors, although a transient acute brain inam-
mation is shown.
75
Gutless vectors have also been used
to successfully express therapeutic genes in the retina
either by transplanting cells previously transduced with
the virus or by direct injection of the vector.
110,111
Other tissues like lung, cardiac muscle, vascular tissue
or dendritic cells have also been targeted with gutless
adenoviruses with similar results as for the muscle or the
liver.
112115
Future considerations
This is a fascinating moment for gutless adenovirus
vectors: their use permits long-term expression in vivo
with reduced and transient cellular immune response in
Gutless adenovirus
R Alba et al
S23
Gene Therapy
animal models for human diseases and, moreover, the
increasing number of groups working in this eld favors
technological advances to escape preimmune response
by developing non-human gutless adenovirus
20
or to
increase stability by generating integrative gutless
vectors,
116,117
or vectors with replication capacity.
118
However, their use in clinical assays is questionable
since helper contamination levels are still high, and
large-scale production in bioreactors is not yet fully
developed. Therefore, future studies will be needed in
these areas to nally bring gutless adenovirus to the
clinic.
Acknowledgements
We would like to acknowledge Dr Merce` Monfar for
critically reading the manuscript. Our work is supported
by MCYT-SAF2003-03256, Marato TV3-2002-031632 and
Instituto de Salud Carlos III (C03/08). AB has a contract
from the Ramon y Cajal Program (Ministerio Educacio n
y Ciencia, Spain), and RA is a recipient of an FI-
Generalitat fellowship.
References
1 Horwitz MS. Adenoviridae and their replication. Virology 1990;
2: 16791720.
2 Danthinne X, Imperiale MJ. Production of rst generation
adenovirus vectors: a review. Gene Therapy 2000; 7: 17071714.
3 Graham FL, Smiley J, Russell WC, Nairn R. Characteristics of a
human cell line transformed by DNA from human adenovirus
type 5. J Gen Virol 1977; 36: 5974.
4 Fallaux FJ et al. Characterization of 911: a new helper cell line
for the titration and propagation of early region 1-deleted
adenoviral vectors. Hum Gene Ther 1996; 7: 215222.
5 Schiedner G, Hertel S, Kochanek S. Efcient transformation of
primary human amniocytes by E1 functions of Ad5: generation
of new cell lines for adenoviral vector production. Hum Gene
Ther 2000; 11: 21052116.
6 Fallaux FJ et al. New helper cells and matched early region
1-deleted adenovirus vectors prevent generation of replication-
competent adenoviruses. Hum Gene Ther 1998; 9: 19091917.
7 Yang Y et al. Cellular immunity to viral antigens limits
E1-deleted adenoviruses for gene therapy. Proc Natl Acad Sci
USA 1994; 91: 44074411.
8 Amaltano A et al. Production and characterization of
improved adenovirus vectors with the E1, E2b, and E3 genes
deleted. J Virol 1998; 72: 926933.
9 Armentano D et al. Effect of the E4 region on the persistence of
transgene expression from adenovirus vectors. J Virol 1997; 71:
24082416.
10 Alemany R et al. Complementation of helper-dependent
adenoviral vectors: size effects and titer uctuations. J Virol
Methods 1997; 68: 147159.
11 Bett AJ, Prevec L, Graham FL. Packaging capacity and stability
of human adenovirus type 5 vectors. J Virol 1993; 67: 59115921.
12 Ghosh-Choudhury G, Graham FL. Stable transfer of a mouse
dihydrofolate reductase gene into a decient cell line using
human adenovirus vector. Biochem Biophys Res Commun 1987;
147: 964973.
13 Mitani K, Graham FL, Caskey CT, Kochanek S. Rescue,
propagation, and partial purication of a helper virus-
dependent adenovirus vector. Proc Natl Acad Sci USA 1995;
92: 38543858.
14 Parks RJ, Graham FL. A helper-dependent system for adeno-
virus vector production helps dene a lower limit for efcient
DNA packaging. J Virol 1997; 71: 32933298.
15 Parks R, Evelegh C, Graham F. Use of helper-dependent
adenoviral vectors of alternative serotypes permits repeat
vector administration. Gene Therapy 1999; 6: 15651573.
16 Schiedner G et al. Variables affecting in vivo performance of
high-capacity adenovirus vectors. J Virol 2002; 76: 16001609.
17 Sandig V et al. Optimization of the helper-dependent adeno-
virus system for production and potency in vivo. Proc Natl Acad
Sci USA 2000; 97: 10021007.
18 Palmer D, Ng P. Improved system for helper-dependent
adenoviral vector production. Mol Ther 2003; 8: 846852.
19 Sato M, Suzuki S, Kubo S, Mitani K. Replication and packaging
of helper-dependent adenoviral vectors. Gene Therapy 2002; 9:
472476.
20 Soudais C, Skander N, Kremer EJ. Long-term in vivo transduc-
tion of neurons throughout the rat CNS using novel helper-
dependent CAV-2 vectors. FASEB J 2004; 18: 391393.
21 Parks RJ et al. A helper-dependent adenovirus vector system:
removal of helper virus by Cre-mediated excision of the viral
packaging signal. Proc Natl Acad Sci USA 1996; 93: 1356513570.
22 Silver DP, Livingston DM. Self-excising retroviral vectors
encoding the Cre recombinase overcome Cre-mediated cellular
toxicity. Mol Cell 2001; 8: 233243.
23 Zang Y, Schneider R. Adenovirus inhibition of cellular protein
synthesis and the specic translation of late viral mRNA. Semin
Virol 1993; 4: 233243.
24 Ng PG, Graham FL. Helper-dependent adenoviral vectors for
gene therapy. In: Templeton NS (ed). Gene and Cell Therapy.
Marcel Dekker Inc: New York, 2004 pp 5370.
25 Som T, Armstrong KA, Volkert FC, Broach JR. Autoregulation
of 2 micron circle gene expression provides a model for
maintenance of stable plasmid copy levels. Cell 1988; 52: 2737.
26 Ng P et al. Development of a FLP/frt system for generating
helper-dependent adenoviral vectors. Mol Ther 2001; 3: 809815.
27 Groth AC, Olivares EC, Thyagarajan B, Calos MP. A phage
integrase directs efcient site-specic integration in human
cells. Proc Natl Acad Sci USA 2000; 97: 59956000.
28 Sargent KL et al. Development of a size-restricted pIX-deleted
helper virus for amplication of helper-dependent adenovirus
vectors. Gene Therapy 2004; 11: 504511.
29 Kubo S, Saeki Y, Chiocca EA, Mitani K. An HSV amplicon-
based helper system for helper-dependent adenoviral vectors.
Biochem Biophys Res Commun 2003; 307: 826830.
30 Cheshenko N, Krougliak N, Eisensmith RC, Krougliak VA. A
novel system for the production of fully deleted adenovirus
vectors that does not require helper adenovirus. Gene Therapy
2001; 8: 846854.
31 Graham FL. Growth of 293 cells in suspension culture. J Gen
Virol 1987; 68 (Pt 3): 937940.
32 Sakhuja K et al. Optimization of the generation and propagation
of gutless adenoviral vectors. Hum Gene Ther 2003; 14: 243254.
33 Schagen FH, Ossevoort M, Toes RE, Hoeben RC. Immune
responses against adenoviral vectors and their transgene
products: a review of strategies for evasion. Crit Rev Oncol
Hematol 2004; 50: 5170.
34 Worgall S, Wolff G, Falck-Pedersen E, Crystal RG. Innate immune
mechanisms dominate elimination of adenoviral vectors follow-
ing in vivo administration. Hum Gene Ther 1997; 8: 3744.
35 Liu Q et al. The role of capsid-endothelial interactions in the
innate immune response to adenovirus vectors. Hum Gene Ther
2003; 14: 627643.
36 Schnell MA et al. Activation of innate immunity in nonhuman
primates following intraportal administration of adenoviral
vectors. Mol Ther 2001; 3: 708722.
37 Molinier-Frenkel V et al. Immune response to recombinant
adenovirus in humans: capsid components from viral input are
Gutless adenovirus
R Alba et al
S24
Gene Therapy
targets for vector-specic cytotoxic T lymphocytes. J Virol 2000;
74: 76787682.
38 Paul WE, Seder RA. Lymphocyte responses and cytokines. Cell
1994; 76: 241251.
39 Kuzmin AI, Finegold MJ, Eisensmith RC. Macrophage deple-
tion increases the safety, efcacy and persistence of adenovirus-
mediated gene transfer in vivo. Gene Therapy 1997; 4: 309316.
40 Wolff G et al. Enhancement of in vivo adenovirus-mediated gene
transfer and expression by prior depletion of tissue macro-
phages in the target organ. J Virol 1997; 71: 624629.
41 Dai Y et al. Cellular and humoral immune responses to
adenoviral vectors containing factor IX gene: tolerization of
factor IX and vector antigens allows for long-term expression.
Proc Natl Acad Sci USA 1995; 92: 14011405.
42 Fang B et al. Gene therapy for hemophilia B: host immunosup-
pression prolongs the therapeutic effect of adenovirus-mediated
factor IX expression. Hum Gene Ther 1995; 6: 10391044.
43 Kaplan JM, Smith AE. Transient immunosuppression with
deoxyspergualin improves longevity of transgene expression
and ability to readminister adenoviral vector to the mouse
lung. Hum Gene Ther 1997; 8: 10951104.
44 Kuriyama S et al. Immunomodulation with FK506 around the
time of intravenous re-administration of an adenoviral vector
facilitates gene transfer into primed rat liver. Int J Cancer 2000;
85: 839844.
45 Otake K, Ennist DL, Harrod K, Trapnell BC. Nonspecic
inammation inhibits adenovirus-mediated pulmonary gene
transfer and expression independent of specic acquired
immune responses. Hum Gene Ther 1998; 9: 22072222.
46 Zuckerman JB et al. A phase I study of adenovirus-mediated
transfer of the human cystic brosis transmembrane conduc-
tance regulator gene to a lung segment of individuals with
cystic brosis. Hum Gene Ther 1999; 10: 29732985.
47 Poller W et al. Stabilization of transgene expression by
incorporation of E3 region genes into an adenoviral factor IX
vector and by transient anti-CD4 treatment of the host. Gene
Therapy 1996; 3: 521530.
48 Sawchuk SJ et al. Anti-T cell receptor monoclonal antibody
prolongs transgene expression following adenovirus-mediated
in vivo gene transfer to mouse synovium. Hum Gene Ther 1996;
7: 499506.
49 Kay MA et al. Long-term hepatic adenovirus-mediated gene
expression in mice following CTLA4Ig administration. Nat
Genet 1995; 11: 191197.
50 Kay MA et al. Transient immunomodulation with anti-CD40
ligand antibody and CTLA4Ig enhances persistence and
secondary adenovirus-mediated gene transfer into mouse liver.
Proc Natl Acad Sci USA 1997; 94: 46864691.
51 Stein CS, Pemberton JL, van Rooijen N, Davidson BL. Effects of
macrophage depletion and anti-CD40 ligand on transgene
expression and redosing with recombinant adenovirus. Gene
Therapy 1998; 5: 431439.
52 Wilson CB et al. Transient inhibition of CD28 and CD40 ligand
interactions prolongs adenovirus-mediated transgene expres-
sion in the lung and facilitates expression after secondary
vector administration. J Virol 1998; 72: 75427550.
53 DeMatteo RP et al. Long-lasting adenovirus transgene expression
in mice through neonatal intrathymic tolerance induction with-
out the use of immunosuppression. J Virol 1997; 71: 53305335.
54 Ilan Y et al. Oral tolerization to adenoviral proteins permits
repeated adenovirus-mediated gene therapy in rats with
pre-existing immunity to adenoviruses. Hepatology 1998; 27:
13681376.
55 Mastrangeli A et al. Sero-switch adenovirus-mediated in vivo
gene transfer: circumvention of anti-adenovirus humoral
immune defenses against repeat adenovirus vector adminis-
tration by changing the adenovirus serotype. Hum Gene Ther
1996; 7: 7987.
56 Morral N et al. Administration of helper-dependent adenoviral
vectors and sequential delivery of different vector serotype for
long-term liver-directed gene transfer in baboons. Proc Natl
Acad Sci USA 1999; 96: 1281612821.
57 Fitzgerald JC et al. A simian replication-defective adenoviral
recombinant vaccine to HIV-1 gag. J Immunol 2003; 170:
14161422.
58 Kremer EJ, Boutin S, Chillon M, Danos O. Canine adenovirus
vectors: an alternative for adenovirus-mediated gene transfer.
J Virol 2000; 74: 505512.
59 Chillon M, Lee JH, Fasbender A, Welsh MJ. Adenovirus
complexed with polyethylene glycol and cationic lipid is
shielded from neutralizing antibodies in vitro. Gene Therapy
1998; 5: 9951002.
60 Croyle MA, Chirmule N, Zhang Y, Wilson JM. Stealth
adenoviruses blunt cell-mediated and humoral immune re-
sponses against the virus and allow for signicant gene
expression upon readministration in the lung. J Virol 2001; 75:
47924801.
61 Croyle MA, Chirmule N, Zhang Y, Wilson JM. PEGylation
of E1-deleted adenovirus vectors allows signicant gene
expression on readministration to liver. Hum Gene Ther 2002;
13: 18871900.
62 Lochmuller H et al. Immunosuppression by FK506 markedly
prolongs expression of adenovirus-delivered transgene in
skeletal muscles of adult dystrophic [mdx] mice. Biochem
Biophys Res Commun 1995; 213: 569574.
63 Vilquin JT et al. FK506 immunosuppression to control the
immune reactions triggered by rst-generation adenovirus-
mediated gene transfer. Hum Gene Ther 1995; 6: 13911401.
64 Chen HH et al. DNA from both high-capacity and rst-
generation adenoviral vectors remains intact in skeletal muscle.
Hum Gene Ther 1999; 10: 365373.
65 Maione D et al. Prolonged expression and effective read-
ministration of erythropoietin delivered with a fully deleted
adenoviral vector. Hum Gene Ther 2000; 11: 859868.
66 Morsy MA et al. An adenoviral vector deleted for all viral
coding sequences results in enhanced safety and extended
expression of a leptin transgene. Proc Natl Acad Sci USA 1998;
95: 78667871.
67 ONeal WK et al. Toxicity associated with repeated adminis-
tration of rst-generation adenovirus vectors does not occur
with a helper-dependent vector. Mol Med 2000; 6: 179195.
68 Schiedner G et al. Genomic DNA transfer with a high-capacity
adenovirus vector results in improved in vivo gene expression
and decreased toxicity. Nat Genet 1998; 18: 180183.
69 Roth MD et al. Helper-dependent adenoviral vectors efciently
express transgenes in human dendritic cells but still stimulate
antiviral immune responses. J Immunol 2002; 169: 46514656.
70 Muruve DA et al. Helper-dependent adenovirus vectors elicit
intact innate but attenuated adaptive host immune responses in
vivo. J Virol 2004; 78: 59665972.
71 De Geest BR, Van Linthout SA, Collen D. Humoral immune
response in mice against a circulating antigen induced by
adenoviral transfer is strictly dependent on expression in
antigen-presenting cells. Blood 2003; 101: 25512556.
72 Brown BD et al. Factors inuencing therapeutic efcacy and the
host immune response to helper-dependent adenoviral gene
therapy in hemophilia A mice. J Thromb Haemost 2004; 2:
111118.
73 Kremer EJ. CAR chasing: canine adenovirus vectors-all bite and
no bark? J Gene Med 2004; 6 (Suppl 1): S139S151.
74 Thomas CE et al. Peripheral infection with adenovirus causes
unexpected long-term brain inammation in animals injected
intracranially with rst-generation, but not with high-capacity,
adenovirus vectors: toward realistic long-term neurological
gene therapy for chronic diseases. Proc Natl Acad Sci USA 2000;
97: 74827487.
Gutless adenovirus
R Alba et al
S25
Gene Therapy
75 Thomas CE et al. Pre-existing antiadenoviral immunity is not a
barrier to efcient and stable transduction of the brain,
mediated by novel high-capacity adenovirus vectors. Hum
Gene Ther 2001; 12: 839846.
76 Brunetti-Pierri N et al. Acute toxicity after high-dose systemic
injection of helper-dependent adenoviral vectors into non-
human primates. Hum Gene Ther 2004; 15: 3546.
77 Croyle MA et al. PEGylated helper-dependent adenoviral
vectors: highly efcient vectors with an enhanced safety prole.
Gene Therapy 2005; 12: 579587.
78 Mok H, Palmer DJ, Ng P, Barry MA. Evaluation of polyethylene
glycol modication of rst-generation and helper-dependent
adenoviral vectors to reduce innate immune responses. Mol
Ther 2005; 11: 6679.
79 Harui A et al. Vaccination with helper-dependent adenovirus
enhances the generation of transgene-specic CTL. Gene
Therapy 2004; 11: 16171626.
80 Chen HH et al. Persistence in muscle of an adenoviral vector
that lacks all viral genes. Proc Natl Acad Sci USA 1997; 94:
16451650.
81 Kim IA et al. Potential of adenoviral p53 gene therapy and
irradiation for the treatment of malignant gliomas. Int J Oncol
2001; 19: 10411047.
82 Morral N et al. High doses of a helper-dependent adenoviral
vector yield supraphysiological levels of alpha1-antitrypsin
with negligible toxicity. Hum Gene Ther 1998; 9: 27092716.
83 Ehrhardt A et al. A gene-deleted adenoviral vector results
in phenotypic correction of canine hemophilia B without
liver toxicity or thrombocytopenia. Blood 2003; 102:
24032411.
84 Reddy PS et al. Sustained human factor VIII expression in
hemophilia A mice following systemic delivery of a gutless
adenoviral vector. Mol Ther 2002; 5: 6373.
85 Belalcazar LM et al. Long-term stable expression of human
apolipoprotein AI mediated by helper-dependent adenovirus
gene transfer inhibits atherosclerosis progression and remodels
atherosclerotic plaques in a mouse model of familial hyper-
cholesterolemia. Circulation 2003; 107: 27262732.
86 Oka K et al. Long-term stable correction of low-density
lipoprotein receptor-decient mice with a helper-dependent
adenoviral vector expressing the very low-density lipoprotein
receptor. Circulation 2001; 103: 12741281.
87 Pastore L et al. Helper-dependent adenoviral vector-mediated
long-term expression of human apolipoprotein AI reduces
atherosclerosis in apo E-decient mice. Gene 2004; 327: 153160.
88 Mian A et al. Long-term correction of ornithine transcarbamy-
lase deciency by WPRE-mediated overexpression using a
helper-dependent adenovirus. Mol Ther 2004; 10: 492499.
89 Kojima H et al. NeuroD-betacellulin gene therapy induces islet
neogenesis in the liver and reverses diabetes in mice. Nat Med
2003; 9: 596603.
90 Aurisicchio L et al. Liver-specic alpha 2 interferon gene
expression results in protection from induced hepatitis. J Virol
2000; 74: 48164823.
91 Fiedler M et al. Helper-dependent adenoviral vector-mediated
delivery of woodchuck-specic genes for alpha interferon (IFN-
alpha) and IFN-gamma: IFN-alpha but not IFN-gamma reduces
woodchuck hepatitis virus replication in chronic infection in
vivo. J Virol 2004; 78: 1011110121.
92 Pastore L et al. Use of a liver-specic promoter reduces immune
response to the transgene in adenoviral vectors. Hum Gene Ther
1999; 10: 17731781.
93 Kass-Eisler A et al. Circumventing the immune response
to adenovirus-mediated gene therapy. Gene Therapy 1996; 3:
154162.
94 Mack CA et al. Circumvention of anti-adenovirus neutralizing
immunity by administration of an adenoviral vector of an
alternate serotype. Hum Gene Ther 1997; 8: 99109.
95 Roy S, Shirley PS, McClelland A, Kaleko M. Circumvention of
immunity to the adenovirus major coat protein hexon. J Virol
1998; 72: 68756879.
96 Smith CA, Woodruff LS, Rooney C, Kitchingman GR. Extensive
cross-reactivity of adenovirus-specic cytotoxic T cells. Hum
Gene Ther 1998; 9: 14191427.
97 Burcin MM et al. Adenovirus-mediated regulable target gene
expression in vivo. Proc Natl Acad Sci USA 1999; 96: 355360.
98 Salucci V et al. Tight control of gene expression by a helper-
dependent adenovirus vector carrying the rtTA2(s)-M2 tetra-
cycline transactivator and repressor system. Gene Therapy 2002;
9: 14151421.
99 Zerby D et al. In vivo ligand-inducible regulation of gene
expression in a gutless adenoviral vector system. Hum Gene
Ther 2003; 14: 749761.
100 Ehrhardt A, Xu H, Kay MA. Episomal persistence of
recombinant adenoviral vector genomes during the cell cycle
in vivo. J Virol 2003; 77: 76897695.
101 Bramson JL et al. Helper-dependent adenoviral vectors contain-
ing modied ber for improved transduction of developing
and mature muscle cells. Hum Gene Ther 2004; 15: 179188.
102 Biermann Vet al. Targeting of high-capacity adenoviral vectors.
Hum Gene Ther 2001; 12: 17571769.
103 Buleld G, Siller WG, Wight PA, Moore KJ. X chromosome-
linked muscular dystrophy (mdx) in the mouse. Proc Natl Acad
Sci USA 1984; 81: 11891192.
104 Dudley RW et al. Sustained improvement of muscle function
one year after full-length dystrophin gene transfer into mdx
mice by a gutted helper-dependent adenoviral vector. Hum
Gene Ther 2004; 15: 145156.
105 Gilbert R et al. Prolonged dystrophin expression and functional
correction of mdx mouse muscle following gene transfer with a
helper-dependent (gutted) adenovirus-encoding murine dys-
trophin. Hum Mol Genet 2003; 12: 12871299.
106 Bilbao R et al. Comparison of high-capacity and rst-generation
adenoviral vector gene delivery to murine muscle in utero.
Gene Therapy 2005; 12: 3947.
107 DelloRusso C et al. Functional correction of adult mdx mouse
muscle using gutted adenoviral vectors expressing full-length
dystrophin. Proc Natl Acad Sci USA 2002; 99: 1297912984.
108 Zou L, Zhou H, Pastore L, Yang K. Prolonged transgene
expression mediated by a helper-dependent adenoviral vector
(hdAd) in the central nervous system. Mol Ther 2000; 2: 105113.
109 Thomas CE et al. Acute direct adenoviral vector cytotoxicity
and chronic, but not acute, inammatory responses correlate
with decreased vector-mediated transgene expression in the
brain. Mol Ther 2001; 3: 3646.
110 Semkova I et al. Autologous transplantation of genetically
modied iris pigment epithelial cells: a promising concept
for the treatment of age-related macular degeneration and
other disorders of the eye. Proc Natl Acad Sci USA 2002; 99:
1309013095.
111 Oshima Y et al. Intraocular gutless adenoviral-vectored VEGF
stimulates anterior segment but not retinal neovascularization.
J Cell Physiol 2004; 199: 399411.
112 Fleury S et al. Helper-dependent adenovirus vectors devoid of
all viral genes cause less myocardial inammation compared
with rst-generation adenovirus vectors. Basic Res Cardiol 2004;
99: 247256.
113 Tuettenberg A et al. Early adenoviral gene expression mediates
immunosuppression by transduced dendritic cell (DC): im-
plications for immunotherapy using genetically modied DC.
J Immunol 2004; 172: 15241530.
114 Wen S, Graf S, Massey PG, Dichek DA. Improved vascular gene
transfer with a helper-dependent adenoviral vector. Circulation
2004; 110: 14841491.
115 Koehler DR et al. Protection of Cftr knockout mice from acute
lung infection by a helper-dependent adenoviral vector
Gutless adenovirus
R Alba et al
S26
Gene Therapy
expressing Cftr in airway epithelia. Proc Natl Acad Sci USA
2003; 100: 1536415369.
116 Goncalves MA et al. Transfer of the full-length dystrophin-
coding sequence into muscle cells by a dual high-capacity
hybrid viral vector with site-specic integration ability. J Virol
2005; 79: 31463162.
117 Yant SR et al. Transposition from a gutless adeno-transposon
vector stabilizes transgene expression in vivo. Nat Biotechnol
2002; 20: 9991005.
118 Kreppel F, Kochanek S. Long-term transgene expression in
proliferating cells mediated by episomally maintained high-
capacity adenovirus vectors. J Virol 2004; 78: 922.
Gutless adenovirus
R Alba et al
S27
Gene Therapy

You might also like