You are on page 1of 11

doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.

com on IDEAL
ARTICLE

Demonstration of Feasibility of In Vivo Gene Therapy


for Gaucher Disease Using a Chemically Induced
Mouse Model
John Marshall,1,* Kerry Anne McEachern,1 Julie A. Cavanagh Kyros,1 Jennifer B. Nietupski,1
Tracey L. Budzinski,1 Robin J. Ziegler,1 Nelson S. Yew,1 Jennifer Sullivan,1 Abraham Scaria,1
Nico van Rooijen,2 John A. Barranger,3 and Seng H. Cheng1
1
Genzyme Corporation, Framingham, Massachusetts 01701-9322, USA
2
Vrije Universiteit, 1081 BT Amsterdam, The Netherlands
3
University of Pittsburgh, Pennsylvania 15261, USA

*
To whom correspondence and reprint requests should be addressed. Fax: (508) 872-4091. E-mail: john.marshall@genzyme.com.

Progress towards developing gene therapy for Gaucher disease has been hindered by the lack
of an animal model. Here we describe a mouse model of Gaucher disease which has a chemically
induced deficiency of glucocerebrosidase and that accumulates elevated levels of glucosylce-
ramide (GL-1) in the lysosomes of Kupffer cells. Administration of mannose-terminated gluco-
cerebrosidase (Cerezyme) resulted in the reduction of GL-1 levels in the livers of these animals.
Gene transduction of hepatocytes with a plasmid DNA vector encoding human glucocerebrosi-
dase (pGZB-GC) generated high-level expression and secretion of the enzyme into systemic cir-
culation with consequent normalization of Kupffer cell GL-1 levels. This suggested that the de
novo synthesized and unmodified enzyme produced by hepatocyte transduction was also capa-
ble of being delivered to the cells that are primarily affected in Gaucher disease.
Immunolocalization studies also revealed that preferential transduction and expression of human
glucocerebrosidase in the Kupffer cells with subsequent reduction in the GL-1 levels could be
attained with a low dose of a recombinant adenoviral vector encoding the human enzyme
(Ad2/CMV-GC). This observation raises the possibility of gene therapy for Gaucher disease that
involves directly transducing the affected histiocytes using recombinant adenoviral vectors.
Together, these data demonstrate the potential for use of in vivo gene therapy vectors for treat-
ing Gaucher disease.

Key Words: Gaucher disease, glucocerebrosidase, glucosylceramide, gene therapy,


recombinant adenoviral vectors, plasmid vectors

INTRODUCTION characterized by neurological disease and by an earlier


Gaucher disease is the most prevalent of the more than age of onset and significantly more severe systemic clin-
40 lysosomal storage disorders that have been described ical symptoms [1].
thus far. It is caused by a deficiency of the lysosomal The involvement of macrophages and the presence of
hydrolase, glucocerebrosidase, resulting in the accumu- a mannose receptor on the surface of these cells provided
lation of the glycolipid glucosylceramide (GL-1) in tissue the basis for enzyme replacement therapy for Gaucher dis-
macrophages [1–3]. Delineation of the three types of ease using glucocerebrosidase containing ␣-mannosyl-ter-
Gaucher disease is based on the severity and age of minated oligosaccharides [4]. Replacement therapy using
disease onset. Type 1, which is the most prevalent, is the such a modified enzyme (Cerezyme) is effective for Type
nonneuropathic form of the disease and is characterized 1 patients but less so for Type 3 and not Type 2 patients
by the accumulation of GL-1 primarily in Kupffer cells, because of the inability of the recombinant enzyme to tra-
osteoclasts, and splenic macrophages. The abnormal verse the blood-brain barrier. Clinical therapy is highly
storage of GL-1 leads to hepatosplenomegaly, pancy- effective but response is heterogenous, with some organs
topenia, lesions in the bone and, in some cases, pul- being less responsive to treatment. Consequently, alter-
monary involvement. Types 2 (acute) and 3 (chronic) are native and adjunctive therapies are also being evaluated,

MOLECULAR THERAPY Vol. 6, No. 2, August 2002 179


Copyright © The American Society of Gene Therapy
1525-0016/02 $35.00
ARTICLE doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL

A P Ι 0.0001
FIG. 1. Localization of GL-1 in the induced Gaucher mouse liver (A). BALB/c
mice were treated with CBE (delivered intraperitoneally) and GL-1-containing
liposomes (delivered intravenously) and 24 hours later, administered with
either clodronate liposomes or PBS. After an additional 24 hours, the livers were
harvested, and the levels of GL-1 in the liver were determined. Treatment with
clodronate liposomes results in depletion of the Kupffer cells. The data shown
are for individual tissue GL-1 values (E) and the mean (▲) ± SEM (n = 6).
Unpaired t-test P values are indicated for the corresponding groups. (B, C, D)
Localization of a fluorescent analog of GL-1. BALB/c mice were injected intra-
venously with GL-1-containing liposomes containing 1% of the fluorescent ana-
log, GL-1B (BODIPY FL C12-glucocerebroside). After 24 hours the livers were
harvested and processed for histological staining. (B) The 5 ␮m sections were
incubated with a rat anti-mouse F4/80 (macrophage marker) antibody and
detected with an AlexaFluor488 goat anti-rat IgG secondary antibody. (C) GL-
1B fluorescence was visualized using a rhodamine filter. The image in (D) is
an overlay of images in (B) and (C), showing co-localization of GL-1B and
macrophages. Arrows depict the co-localization of GL-1B in two separate
macrophages.

B D
rate [15]. These observations suggested that further
improvements in vector design and in the conditions for
transducing human stem cells were necessary.
Evaluation of new therapeutic approaches for Gaucher
disease has been hampered by the lack of a suitable ani-
mal model. Previous attempts to generate either transgenic
knockout mice [16] or mice with targeted mutations akin
to those that cause disease in humans have been unsuc-
C cessful [17–19]. The introduced mutations were all lethal
within hours of birth because of epidermal abnormalities.
These have been characterized as defective development
of the stratum corneum, which results in a compromised
permeability barrier that leads to death by dehydration
[20]. An alternative is an artificially induced model in
which glucocerebrosidase is specifically and irreversibly
inhibited by conduritol B epoxide (CBE), and the glycol-
ipid levels in the Kupffer cells increased by administering
such as using bisphosphonates to treat osteopenic bone GL-1 [21–27]. Mice treated with CBE and GL-1-containing
disease [5], glucosylceramide synthase inhibitors to abate liposomes lack glucocerebrosidase activity [24] and display
the accumulation of GL-1 [6,7], and cell and gene [8–13] elevated levels of the glycolipid, primarily in Kupffer cells
therapies to augment the levels of glucocerebrosidase. Here [25]. As such, these animals display one of the major
we report our efforts to evaluate the feasibility of gene pathological lesions observed in Gaucher disease.
therapy for Gaucher disease. To evaluate the efficacy of alternative gene therapy
The characteristics of the pathobiology of Gaucher approaches for Gaucher disease, we validated the utility
disease make it a logical candidate for gene therapy of the artificially induced murine model. Using this
strategies that are directed at correcting the enzyme defi- model, we asked if direct gene transduction of the liver
ciency in macrophages. Indeed, the majority of the early could act as a “depot” of recombinant glucocerebrosi-
gene therapy efforts for Gaucher disease were focused on dase and if the secreted enzyme cleared GL-1 from
retroviral-mediated transduction of bone marrow stem Kupffer cells. Unlike Cerezyme, gene therapy-derived glu-
cells or peripheral blood monocytes [10–13]. Preclinical cocerebrosidase will not be further modified to expose
studies in mice showed that high-efficiency gene trans- the mannose residues on the oligosaccharide side chains.
fer and stable engraftment of gene-modified bone mar- It was, therefore, important to determine whether or not
row stem cells could be achieved [10,14]. However, sub- the gene therapy-derived enzyme was delivered to the
sequent clinical studies using autologous stem cells from macrophages. Finally, since a proportion of recombinant
Gaucher patients showed that the efficiency of retrovi- adenoviral vectors delivered systemically is internalized
ral transduction of these cells was markedly lower, and by macrophages [28], we investigated whether or not
that engraftment of the modified stem cells in the clearance of GL-1 levels could be realized using a low
absence of ablation was transient and occurred at a low dose of the viral vector.

180 MOLECULAR THERAPY Vol. 6, No. 2, August 2002


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL
ARTICLE

FIG. 2. Effect of adding exogenous recombinant glucocere-


brosidase on GL-1 levels in the induced Gaucher mouse liver.
P = 0.05
BALB/c mice were treated with CBE (delivered intraperi-
toneally) and GL-1-containing liposomes (delivered intra-
venously) and 24 hours later administered either 40 U/kg
Cerezyme or PBS. After an additional 24 hours the livers were
harvested and the levels of GL-1 determined. The data shown
are for individual tissue GL-1 values (O) and the mean (▲) ±
SEM (n = 5). Unpaired t-test P values are indicated for the cor-
responding groups.

GL-1 (GL-1B) were injected intravenously into


CBE-treated mice and the livers were harvested
24 hours later. Histological sections (5 ␮m) of
the livers were probed with two rat anti-mouse
macrophage antibodies (MoMa-1 and F4/80)
followed by a goat anti-rat AlexaFluor350 sec-
ondary antibody. Sections were viewed by flu-
Cerezyme orescence microscopy using Ex503/Em512 for
the GL-1B and Ex346/Em442 for the
AlexaFluor350. All of the GL-1B fluorescence
RESULTS coincided with the AlexaFluor350 fluorescence, indicat-
ing that the GL-1B was localized in the Kupffer cells (Fig.
Characterization of a Chemically Induced Murine 1B). Together, these results demonstrate that treatment
Model of Gaucher Disease with CBE and GL-1-containing liposomes leads to elevated
An evaluation of the feasibility of gene therapy for levels of GL-1 in the Kupffer cells of mice for up to 5 days.
Gaucher disease would benefit from the availability of an Histiocyte storage of GL-1 is one of the hallmarks of
animal model. Because mice whose gene for glucocere- Gaucher disease, and these animals represent a working
brosidase has been disrupted by insertional mutagenesis model to evaluate the feasibility of gene therapy for treat-
are nonviable [16], we elected to adapt an induced ing this disease.
Gaucher mouse model that was first described by Kanfer To confirm that the GL-1 delivered to the Kupffer cells
et al. [21]. In this model, the animals are treated with CBE of the induced Gaucher mouse model was located within
to deplete endogenous enzyme activity and liposomes con- the lysosomal compartment, the mice were injected intra-
taining GL-1 to augment the accumulation of the gly- venously with 40 U/kg Cerezyme. Cerezyme is a recombi-
cosphingolipid in the lysosomes of Kupffer cells. Following nant form of glucocerebrosidase in which the oligosac-
treatment with CBE and GL-1-containing liposomes, the charide side chains have been enzymatically modified to
levels of glucocerebrosidase in the livers were reduced by expose terminal mannose residues. Modification of the
greater than 90% and the levels of GL-1 were increased enzyme in this manner facilitates preferential targeting of
5- to 10-fold over those observed in normal mice (~ 5 ␮g the enzyme to the mannose receptors on the macrophages
GL/g tissue) (Fig. 1A). These elevated levels of GL-1 in the [4]. Since glucocerebrosidase is only active at a pH less
induced mouse liver were sustained for up to 5 days post- than 6, a reduction in GL-1 levels in the liver of the
treatment (data not shown). induced Gaucher mouse would indicate that the glycol-
As Gaucher disease manifests in the liver as a result of ipids were located in the lysosomes. Treatment with
accumulation of GL-1 primarily in the Kupffer cells, we Cerezyme resulted in a reduction of the GL-1 levels in the
asked whether or not the elevated levels of GL-1 in the arti- livers of the induced Gaucher mice compared to those of
ficially-induced model of Gaucher disease were due to naïve animals (Fig. 2). For the GL-1 to be hydrolyzed by
accumulation of the glycosphingolipid in this cell type. To Cerezyme, most of the glycolipid delivered to the Kupffer
test this, mice that had been administered CBE and GL-1- cells of the induced Gaucher mouse must consequently
containing liposomes were treated 24 hours later with clo- have been localized within the acidic lysosomal compart-
dronate-encapsulated liposomes to transiently deplete the ment.
liver of macrophages [29,30]. Treatment with clodronate
liposomes reduced the elevated levels of GL-1 to basal lev- Cellular Localization of Modified Cerezyme and
els (Fig. 1A), indicating that the majority of the liver accu- Unmodified Recombinant Human Glucocerebrosidase
mulation of GL-1 in the induced Gaucher mouse was in in the Liver
the Kupffer cells. This result was confirmed using a fluo- Enzyme replacement therapy using Cerezyme has been
rescent analog of GL-1 (BODIPYFL-tagged C12-glucocere- shown to be efficacious in the treatment of Gaucher Type
broside). Liposomes containing this fluorescently labeled 1 patients [31]. This suggests that gene transduction of an

MOLECULAR THERAPY Vol. 6, No. 2, August 2002 181


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL

FIG. 3. Localization of systemically deliv-


A B C ered recombinant human glucocerebrosi-
dase in the liver. Fischer 344 rats were
administered intravenously with 80 U/kg
Cerezyme (A–F) or with 80 U/kg unmodi-
fied glucocerebrosidase (G–I). The livers
were harvested 1 hour later and processed
for histological staining. Liver sections were
incubated with two anti-human gluco-
cerebrosidase monoclonal antibodies (1B5
and 8E4) and detected with an
AlexaFluor568 goat anti-mouse IgG1 sec-
ondary antibody (A, D, G). The same sec-
tions were also incubated with either a
mouse anti-rat CD11b (macrophage
D E F marker) antibody (B, H), or a sheep anti-rat
von Willebrand factor (endothelial cell
marker) antibody (E). These were detected
using an Alexafluor488 goat anti-mouse
IgG2a and an AlexaFluor488 donkey anti-
sheep secondary antibody, respectively.
Images in (C), (F), and (I) are overlays of the
corresponding images from (A), (D), and
(G) with (B), (E) and (H), respectively.
Arrows indicate examples of cells that show
human glucocerebrosidase localized in
either the macrophages (C, I) or endothe-
lial cells (F).

G H I

appropriate depot organ with subsequent secretion of glu- cell morphology, very little of the enzyme appeared to be
cocerebrosidase could also be effective. However, as indi- localized in the hepatocytes. The same liver sections were
cated earlier, Cerezyme is a modified form of glucocere- costained with either CD11b, a macrophage-specific anti-
brosidase in which the oligosaccharides have been altered body (Fig. 3B), or with an anti-rat von Willebrand factor
to improve delivery to the Kupffer cells. Since glucocere- antibody, an endothelial cell marker (Fig. 3E). Figure 3C
brosidase produced following gene therapy is unlikely to is an overlay of the images from Figs. 3A and 3B showing
be similarly processed, it was important to determine co-localization of Cerezyme in a number of the
whether unmodified enzyme also localized to the Kupffer macrophages (shown as yellow cells), and Figure 3F is an
cells. overlay of Figs. 3D and 3E showing that the majority of
To address this question, Cerezyme and unmodified the endothelial cells had internalized Cerezyme. These
recombinant glucocerebrosidase (purified from CHO cells) data indicate that, following intravenous infusion of
were localized in rat livers following intravenous infusion. Cerezyme, most of the enzyme is internalized by endothe-
Animals were administered 80 U/kg Cerezyme and the liv- lial cells, some by macrophages, and very little by hepa-
ers were harvested 1 hour later and processed for histo- tocytes. This biodistribution of Cerezyme in the different
logical analysis. The liver sections were probed with anti- liver cell types is concordant with published data [32].
bodies specific for human glucocerebrosidase, This experiment was repeated using unmodified recom-
macrophages, and endothelial cells. Figures 3A and 3D are binant human glucocerebrosidase that had been purified
representative liver sections stained with antibodies spe- from CHO cells. To determine if unmodified enzyme could
cific for human glucocerebrosidase showing abundant be internalized by macrophages, liver sections of rats treated
staining of the enzyme in these tissue sections. Based on with 80 U/kg unmodified recombinant glucocerebrosidase

182 MOLECULAR THERAPY Vol. 6, No. 2, August 2002


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL
ARTICLE

Ad2/CMV-GC
and supports the feasibility of gene therapy for treating
Gaucher disease.

Efficacy of Adenoviral-Mediated Gene Therapy of


Gaucher Disease
To extend these observations to gene therapy-derived
human glucocerebrosidase, a recombinant adenoviral vec-
tor encoding human glucocerebrosidase (Ad2/CMV-GC)
was generated. Intravenous administration of 5 ⫻ 1012 par-
ticles/kg of Ad2/CMV-GC into BALB/c mice resulted in
high-level expression of glucocerebrosidase in the liver
(100-fold endogenous levels), spleen (10-fold endogenous
levels), and lungs (10-fold endogenous levels) as well as
FIG. 4. Tissue expression levels of glucocerebrosidase following systemic deliv- secretion of significant levels into the serum (Fig. 4). The
ery of the recombinant adenoviral vector Ad2/CMV-GC. BALB/c mice received level of glucocerebrosidase attained in the serum was deter-
an intravenous injection of 5 ⫻ 1012 particles/kg of Ad2/CMV-GC. Serum,
mined to be approximately 10-fold higher than the serum
liver, spleen, and lungs were analyzed for glucocerebrosidase activity 2 days
post-injection. Levels of the enzyme in naïve BALB/c mice tissues were also steady state levels of Cerezyme in Gaucher patients dur-
assayed. The dashed line indicates the steady state concentration of Cerezyme ing a therapeutic infusion (Fig. 4). This suggests that gene
attained in the serum of Gaucher patients during a therapeutic infusion. Data transduction using a high dose of Ad2/CMV-GC can result
shown are expressed as mean ± SEM (n = 8). in the production of therapeutic levels of glucocerebrosi-
dase in the serum as well as supraphysiological levels of
enzyme in various Gaucher-affected tissues.
and processed 1 hour later were stained for the enzyme (Fig. The distribution of human glucocerebrosidase among
3G) and for macrophages (Fig. 3H). The staining pattern for the different liver cell types following treatment with
the unmodified glucocerebrosidase (Fig. 3G) was similar to Ad2/CMV-GC was also examined. Rats were administered
that for Cerezyme (Figs. 3A and 3D), except that some of either 8 ⫻ 1012 (Figs. 5A–5C) or 2 ⫻ 1012 (Figs. 5D–5F) par-
the hepatocytes also stained positive. Figure 3I is an over- ticles/kg of Ad2/CMV-GC, and the livers were examined 2
lay of 3G and 3H showing the localization of unmodified days later. Immunofluorescence staining revealed that the
glucocerebrosidase in some of the macrophages (shown as majority of the human glucocerebrosidase in the livers of
yellow cells). This result indicates that unmodified gluco- rats that received the higher dose of the recombinant ade-
cerebrosidase retains the ability to localize to Kupffer cells novirus vector was in the hepatocytes (Fig. 5A).

A B C

FIG. 5. Localization of recombinant human


glucocerebrosidase in the liver following sys-
temic delivery of Ad2/CMV-GC. Fischer 344
rats were administered intravenously with
either 8 ⫻ 1012 particles/kg (A–C) or 2 ⫻
1012 particles/kg (D–F) Ad2/CMV-GC. The
livers were processed for histological stain-
ing2 days post-injection. Liver sections were
incubated with two anti-human glucocere-
brosidase monoclonal antibodies (1B5 and D E F
8E4) and detected with an Alexafluor568
goat anti-mouse IgG1 secondary antibody
(A, D). The same sections were also incu-
bated with a mouse anti-rat CD11b
(macrophage marker) antibody (B, E), and
detected using AlexaFluor488 goat anti-
mouse IgG2a. The image in (C) is an overlay
of the images in (A) and (B), and the image
in (F) is an overlay of the images in (D) and
(E). Arrows indicate examples of cells show-
ing both human glucocerebrosidase and
macrophage staining.

MOLECULAR THERAPY Vol. 6, No. 2, August 2002 183


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL

in GL-1 was most likely a consequence of viral


P = 0.001
transduction of the affected Kupffer cells.
Hence, administration of a low dose of a
recombinant adenoviral vector encoding glu-
cocerebrosidase may be efficacious in reduc-
ing the lysosomal storage abnormality in the
liver.

Ability of Kupffer Cells to Internalize


Recombinant Glucocerebrosidase Secreted
from Genetically Modified Hepatocytes
Because recombinant adenoviral vectors
directly transduced the Kupffer cells, we were
unable to surmise whether glucocerebrosidase
secreted from the transduced hepatocytes was
able to cross-correct GL-1 accumulation in the
Ad2/CMV-GC Kupffer cells. To address this question, a plas-
mid DNA vector encoding human glucocere-
brosidase (pGZB-sGC) was constructed and
FIG. 6. Effect of intravenous administration of Ad2/CMV-GC on liver GL-1 levels in the delivered hydrodynamically into the livers of
induced Gaucher mouse. BALB/c mice were treated with CBE (delivered intraperitoneally) and mice. This procedure results in the transduc-
GL-1-containing liposomes (delivered intravenously) and 24 hours later administered either 5 tion of 5–10% of hepatocytes and less than
⫻ 106, 1.5 ⫻ 106, or 0.5 ⫻ 1012 particles/kg Ad2/CMV-GC. The animals were killed, and the lev-
els of GL-1 in their livers were assayed 2 days later. The data shown are for individual tissue GL-
0.1% of Kupffer cells (data not shown)
1 values (E) and the mean (F) ± SEM (n = 6). Unpaired t-test P values are indicated for the cor- [35,36]. Hydrodynamic injection of 10 ␮g of
responding groups. pGZB-sGC into the liver of the induced
Gaucher mice resulted in an approximately
fourfold higher level of glucocerebrosidase in
Superimposing the image of this section with that stained the liver and an approximately 20-fold higher level in the
for macrophages (Fig. 5B) revealed co-localization (shown serum than that observed in untreated mice (Fig. 7A).
as yellow cells in Fig. 5C) in 10–20% of the Kupffer cells. Concomitant with these levels of expression was a reduc-
Rats treated with the lower dose of Ad2/CMV-GC (2 ⫻ 1012 tion in the GL-1 content in the livers of these animals to
particles/kg) showed little evidence of glucocerebrosidase basal levels (Fig. 7B). That the reduction in GL-1 was due
in the parenchymal cells (Fig. 5D). Instead, the majority to expression of glucocerebrosidase, rather than to the pro-
of the enzyme was localized in the Kupffer cells (Fig. 5F). cedure used to transfect the liver, was demonstrated by the
These observations are consistent with the notion that a finding that mice treated with an irrelevant plasmid
proportion of adenoviral vectors administered systemi- (pGZB-s␣Gal, a plasmid DNA vector encoding another
cally are sequestered by the reticuloendothelial cells, and lysosomal enzyme, human ␣-galactosidase A) did not
that administration of doses above this threshold results affect GL-1 levels. Since hydrodynamic-mediated trans-
in the additional transduction of hepatocytes [28,33,34]. duction resulted in expression of glucocerebrosidase pri-
This pattern of transgene expression suggests that gene marily in the hepatocytes, our finding of complete clear-
therapy using a relatively low dose of Ad2/CMV-GC may ance of GL-1 strongly suggests that the secreted enzyme
suffice to reverse the accumulation of GL-1 in histiocytes was susceptible to uptake by the neighboring and possi-
that are primarily affected in Gaucher disease. bly the distant Kupffer cells. Hence, gene delivery vectors
To determine if administration of low doses of recom- that effect genetic modification of depot organs with sub-
binant adenoviral vectors could be therapeutic, a dose- sequent secretion of unmodified glucocerebrosidase
response study was performed in the induced Gaucher should be efficacious in treating Gaucher disease.
mouse model. Mice treated with CBE and GL-1-contain-
ing liposomes were infused increasing amounts of Clearance of GL-1 from Kupffer Cells Following
Ad2/CMV-GC ranging between 0.5 and 5 ⫻ 1012 parti- Intranasal Instillation of Ad2/CMV-GC
cles/kg. Analysis of the livers of these mice 2 days later To further demonstrate that glucocerebrosidase secreted
showed that a low dose of Ad2/CMV-GC (0.5 ⫻ 1012 par- from gene-transduced cells is capable of being internalized
ticles/kg) was equally effective at clearing the GL-1 from by Kupffer cells, mice were instilled intranasally with 5 ⫻
the Kupffer cells as the higher doses (Fig. 6). Since mice 1011 particles/kg of Ad2/CMV-GC. It has been shown that
treated with 0.5 ⫻ 1012 particles/kg of Ad2/CMV-GC did pulmonary instillation of a recombinant adenoviral vector
not result in secretion of detectable levels of glucocere- encoding the lysosomal enzyme ␣-galactosidase A results
brosidase into the serum (data not shown), the reduction in transduction of the lung with subsequent secretion of

184 MOLECULAR THERAPY Vol. 6, No. 2, August 2002


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL
ARTICLE

A FIG. 7. Plasmid DNA-mediated transduction of the livers of induced


Gaucher mice. (A) Tissue expression levels of glucocerebrosidase following
pGZB-sGC
hydrodynamic delivery of the plasmid pGZB-sGC. BALB/c mice were treated
with CBE (delivered intraperitoneally) and GL-1-containing liposomes (deliv-
ered intravenously) and then 24 hours later, injected hydrodynamically with
10 ␮g of pGZB-sGC. The levels of glucocerebrosidase were assayed in the
serum and liver 2 days post-injection. The levels of glucocerebrosidase in naïve
mice and in mice injected with pGZB-␣gal (encoding human ␣-galactosidase
A) were also determined. Data shown are expressed as mean ± SEM (n = 8).
(B) Effect of hydrodynamic administration of pGZB-sGC on GL-1 levels in the
induced Gaucher mouse. Mice were treated as described above. The levels of
GL-1 in the livers were assayed 2 days later. The data shown are for individ-
ual tissue GL-1 values (O) and the mean (▲) ± SEM (n = 8). Unpaired t-test P
values are indicated for the corresponding groups.

has been modified to facilitate preferential targeting to


B macrophages results in the clearance of accumulated GL-
P = 0.001 1 from the affected organs with consequent clinical
improvement. However, because the half-life of gluco-
cerebrosidase is relatively short, sustained clinical improve-
ment requires biweekly infusions of the recombinant
P Ι 0.001
enzyme for life. Because gene therapy provides the possi-
bility for prolonged expression of the enzyme, we have
investigated the feasibility of this approach as an alterna-
tive modality for treating Gaucher disease.
Much of the early gene therapy effort for Gaucher dis-
ease was directed at evaluating ex vivo approaches [10–13].
Because the clinical experience using this approach has
been unsatisfactory [15], the focus of the current studies
was centered around the use of in vivo gene delivery vec-
pGZB-sGC tors. Gene therapy for Gaucher disease using in vivo gene
transfer vectors poses several technical challenges. Firstly,
glucocerebrosidase is not normally secreted unless the cells
the enzyme into systemic circulation [37]. Since no viral express very high levels of the enzyme. As such, the gene
vector was detected outside of the lung following this route delivery vectors chosen need to be extremely efficient in
of delivery, the detection of enzyme in other visceral organs their ability to effect expression of the enzyme. Secondly,
indicated that enzyme was secreted from the lung and rein- unlike the majority of lysosomal enzymes, glucocere-
ternalized by these other tissues. Figure 8 shows that mice brosidase is targeted to the lysosomes by way of a man-
administered intranasally with Ad2/CMV-GC can effect a nose-6-phosphate-independent pathway. The exact mech-
reduction of GL-1 in the Kupffer cells of the chemically anism by which the enzyme is delivered to the lysosomal
induced Gaucher mouse. This suggests that glucocere- compartment is still unclear. Efficient targeting of recom-
brosidase was similarly secreted from the transduced lung binant glucocerebrosidase to the affected macrophages is
into systemic circulation and confirms that the glucocere- currently realized by modifying the purified enzyme to
brosidase produced by gene transfer can be delivered effec- expose mannose residues on the oligosaccharides as is the
tively to Kupffer cells. case with Cerezyme. However, this additional processing
is untenable with glucocerebrosidase that is produced by
gene therapy. Therefore, it was important to determine if
DISCUSSION de novo synthesized and secreted glucocerebrosidase
The primary pathological manifestation of Gaucher disease obtained following in vivo gene transduction could be
is the accumulation of GL-1 in the lysosomes of tissue internalized by tissue macrophages. Finally, progress
macrophages resulting in enlarged, so-called “Gaucher toward evaluating novel therapeutic approaches for
cells” [1–3]. The resultant perturbation causes significant Gaucher disease has been hindered by the absence of a
enlargement of the liver and spleen, as well as other tis- viable animal model in which to test efficacy.
sue defects such as pulmonary complications and bone In this report, we validated the utility of an induced
lesions. The nonneuropathic form of Gaucher disease can mouse model of Gaucher disease. The generation of such
be treated successfully with enzyme replacement therapy a model with accumulation of GL-1 in the liver has been
[31]. Therapy using recombinant glucocerebrosidase that described [21,25], though there are no reports of this

MOLECULAR THERAPY Vol. 6, No. 2, August 2002 185


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL

P = 0.001
GL-1 could be hydrolyzed readily following a
bolus intravenous injection of Cerezyme. This
enzyme is weakly active at neutral pH and only
functions optimally in the acidic environment
of a lysosome. We surmise that Cerezyme must
have accumulated within the lysosomes of the
tissue macrophages for treatment with it to
lower the liver GL-1 levels. This demonstration
also helped validate the induced mouse as a
suitable model of Gaucher disease because it
was successfully treated with the same thera-
peutic used for Gaucher patients.
Using the chemically induced Gaucher
mouse, studies were performed to examine the
feasibility of gene transfer for treating this dis-
ease. Gene transduction using either recombi-
Ad2/CMV-GC IV nant adenoviral or plasmid DNA vectors
Ad2/CMV-GC IN resulted in expression and secretion of thera-
peutic levels of glucocerebrosidase into the
FIG. 8. Effect of intranasal delivery of Ad2/CMV-GC on liver GL-1 levels in the induced Gaucher
mouse. BALB/c mice were treated with CBE (delivered intraperitoneally) and GL-1-containing
serum. Moreover, levels of the enzyme were
liposomes (delivered intravenously) and 24 hours later administered 5 ⫻ 1011 particles/kg also detected in the liver, spleen and lung, the
Ad2/CMV-GC either intravenously (i.v.) or intranasally (i.n.). The animals were killed and the organs that are primarily affected in Gaucher
levels of GL-1 in the livers were assayed 2 days later. The data shown are for individual tissue disease. Systemic delivery of 5⫻1012 particles/kg
GL-1 values (O) and the mean (▲) ± SEM (n = 6). Unpaired t-test P values are indicated for
of Ad2/CMV-GC generated levels of glucocere-
the corresponding groups.
brosidase in the serum that are predicted to be
therapeutic with consequent clearance of GL-1
from the livers of the induced Gaucher mouse.
model being used to evaluate therapeutics. Model valida- Interestingly, low doses of the recombinant adenoviral
tion was achieved by demonstrating that the elevated lev- vector were equally effective at reducing GL-1 levels, pre-
els of the GL-1 in the liver were primarily associated with sumably because the virus transduced the macrophages
Kupffer cells, localized within the lysosomal compartment, following uptake [28]. This observation suggests that a low
and metabolically accessible. That the GL-1 was located in dose of a recombinant adenoviral vector may be effica-
the liver macrophages was demonstrated by treating the cious at reducing the storage abnormality in the liver.
animals with clodronate-encapsulated liposomes [29] and However, because this dose did not result in secretion of
by showing a concomitant loss in liver GL-1 levels. This the enzyme, it is unlikely that it would alter the GL-1
conclusion was further verified by immunolocalization accumulation in other affected histiocytes that were not
studies showing that fluorescently tagged GL-1 was pri- transduced by the adenoviral vector. Thus, use of low doses
marily located in the Kupffer cells. These observations are of adenoviral vectors may have limited utility.
congruent with those reported earlier showing that GL-1 Because the primary affected cells in Gaucher disease
delivered as an emulsion or liposomal formulation results are the tissue macrophages, a key question was whether
primarily in liver deposition, with approximately equal or not gene transfer-derived glucocerebrosidase containing
amounts going to hepatocytes and Kupffer cells [25–27]. unmodified oligosaccharide side chains could be inter-
However, the GL-1 in the hepatocytes is lost within 24 nalized by these cells. Systemic administration of recom-
hours through biliary excretion and the remaining levels binant adenoviral vectors resulted in the delivery of a pro-
of liver GL-1, which are relatively stable for several days, portion of the virus to the macrophages; thus, we were
are associated primarily with the Kupffer cells [26,38]. It unable to address this issue using this viral vector. We,
should be noted that the GL-1 used in these studies was therefore, employed pressure-mediated delivery of plas-
derived from plants. However, this material contains a mid DNA encoding glucocerebrosidase to the liver. The
C-16 ceramide, which is one of the more common forms results demonstrated that unmodified glucocerebrosidase
of GL-1 derived from the visceral organs of Gaucher generated by hydrodynamic-mediated delivery of pGZB-
patients. sGC reduced the levels of GL-1 in the Kupffer cells. Because
We also demonstrated that the GL-1 in the induced hydrodynamic-mediated delivery of plasmid DNA results
Gaucher mouse was appropriately stored within the lyso- primarily in transduction of hepatocytes [35,36], the com-
somes of the Kupffer cells and that it was accessible for plete clearance of GL-1 strongly suggested that low levels
enzymatic hydrolysis. The subcellular localization was of the enzyme secreted from the hepatocytes were taken
determined indirectly by demonstrating that the stored up by the macrophages. That glucocerebrosidase produced

186 MOLECULAR THERAPY Vol. 6, No. 2, August 2002


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL
ARTICLE

by gene transfer can be targeted to Kupffer cells was veri- of GL-1B used in the formulations was approximately 1% (by mass) of the
fied by the observation that Ad2/CMV-GC delivered total GL-1.
intranasally into the lung also resulted in a reduction of Generating an artificially induced Gaucher mouse model. All procedures
GL-1 levels in the Kupffer cells. In this instance, the involving animals were reviewed and approved by an Institutional Animal
Care and Use Committee (IACUC) following Association for Assessment of
enzyme was produced in the lung and secreted into sys- Accreditation of Laboratory Animal Care (AAALAC), State and Federal
temic circulation. The notion that the unmodified gluco- guidelines. To generate the induced Gaucher mouse model, female BALB/c
cerebrosidase generated by gene transfer could be inter- mice (6–8 weeks old; Taconic, Germantown, NY) were first injected
nalized by macrophages in vivo was further supported by intraperitoneally with 100 mg/kg CBE (Sigma, St. Louis, MO) as described
[22]. The mice were challenged either intraperitoneally with an emulsion
studies using purified recombinant enzyme showing sim-
of GL-1 (GL-1:Myrj-52 emulsion; 500 mg GL-1/kg body weight) or intra-
ilar uptake, albeit at only 10–20% the efficiency of man- venously into the tail vein with a liposomal formulation of GL-1 (PC:PS:GL-
nose-terminated enzyme [39–41]. However, it should be 1 liposomes; 50 mg GL-1/kg body weight) 1 hour later. It has been shown
noted that unmodified glucocerebrosidase is reportedly that injection of CBE results in the complete inhibition of glucocerebrosi-
less labile in serum and more efficiently delivered to the dase activity in all tissues within 1 hour and that the inhibitor is cleared
rapidly (in approximately 5 hours) from the animals through hydrolysis
lysosomal compartment, and exhibits a longer half-life [21,22,24]. As such, CBE should not have any effect on the exogenously
than the oligosaccharide-remodeled enzyme. Thus, administered glucocerebrosidase or on the enzyme produced by gene trans-
although less unmodified enzyme may be delivered to fer.
macrophages, it acts for a longer time. The observation Transient depletion of Kupffer cells. Transient depletion of tissue
that gene transfer-derived glucocerebrosidase could be macrophages was achieved by intravenous administration of clodronate
taken up by macrophages is consistent with previous (gift from Roche Diagnostics) encapsulated in liposomes (clodronate lipo-
somes) as described [29,30]. Liposomes containing clodronate (5 mg/ml)
reports showing that enzyme derived from gene-modified were injected in a total volume of 200 ␮l. Liver sections taken 24 hours
myoblasts contain high mannose-type oligosaccharide after treatment with clodronate liposomes and analyzed by immunohis-
chains [42]. Together, these observations indicate that tochemical staining confirmed that greater than 90% of the macrophages
gene therapy strategies that result in the transduction of in the liver had been eliminated [29].
depot organs with a resultant secretion of glucocerebrosi- Recombinant glucocerebrosidase. Recombinant human glucocerebrosi-
dase should be efficacious in the treatment of Gaucher dase was purified from the media of Chinese hamster ovary cells that had
been stably transfected with an expression vector encoding the human
disease. enzyme. We used two forms of the enzyme: unmodified glucocerebrosidase,
We have validated the utility of a chemically induced and Cerezyme (Genzyme Corp, Cambridge, MA), which is glucocerebrosi-
mouse model of Gaucher disease and showed that it is a dase modified by exoglycosidases to generate mannose-terminated oligosac-
valuable surrogate for the evaluation of therapeutic charide side chains. This modification imparts more efficient targeting of
the enzyme to Kupffer cells and other tissue macrophages by way of man-
approaches. Moreover, gene transduction using recombi-
nose receptors on the cell surface [4]. The purified enzymes (40 U/kg in 5%
nant adenoviral and plasmid DNA vectors can result in the mannitol) were administered into the tail veins of mice as a 100 ␮l bolus
expression and secretion of therapeutic levels of gluco- injections, or to rats (80 U/kg) as 500 ␮l bolus injections.
cerebrosidase. Importantly, we demonstrate that the Gene delivery vectors. The recombinant adenoviral vector, Ad2/CMV-GC
secreted enzyme is in a form that is compatible with encoding human glucocerebrosidase was constructed essentially as
uptake by macrophages, the cell type that is primarily described [43]. Briefly, the E1 region of adenovirus serotype 2 was replaced
with an expression cassette consisting of the human cytomegalovirus
affected in Gaucher disease. We have demonstrated the
(CMV) immediate early promoter and enhancer, the human glucocere-
potential of in vivo gene transfer approaches for treating brosidase cDNA and a simian virus 40 small t polyadenylation signal
this disease. sequence. For intravenous delivery, the recombinant adenoviral vectors
were administered as a 100 ␮l bolus injection into the tail vein in an excip-
ient of 5% sucrose in phosphate-buffered saline (PBS). For pulmonary deliv-
MATERIALS AND METHODS ery, a 100 ␮l volume of the viral vector was instilled intranasally through
Formulation of GL-1-containing emulsions and liposomes. To generate the nares by inspiration.
an emulsion of glucosylceramide (Matreya, Inc., Pleasant Gap, PA), the The plasmid DNA vector, pGZB-sGC encoding a synthetic human glu-
glycolipid was formulated with the detergent Myrj-52 (Sigma, St. Louis, cocerebrosidase was generated as described [44]. The vector was modified
MO) at a ratio of 4:3 (w/w) as described [25]. Briefly, GL-1 was first evap- to contain a minimum number of CpG motifs to improve its toxicity pro-
orated from chloroform and a saline solution (145 mM sodium chloride file. pGZB-sGC contains a minimal origin of replication, a kanamycin resist-
solution) of Myrj-52 was then added to achieve a final glycolipid concen- ance gene and an expression cassette composed of the CMV promoter and
tration of 20 mg/ml. The mixture was sonicated using a probe sonicator enhancer, a hybrid intron, the human glucocerebrosidase cDNA (that had
(VirSonic 100, Virtis Inc., Gardiner, NY) until an even dispersion was been codon optimized for the most abundant mammalian tRNAs), and a
achieved. To generate the liposomal formulation, glucosylceramide (GL-1) bovine growth hormone polyadenylation signal sequence. The plasmid
was formulated with phosphatidylcholine (PC) and phosphatidylserine DNA vector, pGZB-saGal is identical to pGZB-sGC except that the gluco-
(PS) at a ratio of 1:8:1 (w/w/w). Chloroform solutions of the appropriate cerebrosidase cDNA was replaced with that for human ␣-galactosidase A.
ratios of PC, PS and GL-1 were first mixed and then evaporated to dryness Hydrodynamic delivery of the plasmid DNA vectors was performed using
on a rotary evaporator. The dried lipid film was then hydrated in saline the TransIT in vivo gene delivery system (Mirus Corp., Madison, WI) as out-
(GL-1 final concentration 5mg/ml) using a bath sonicator until an even sus- lined by the manufacturer and essentially as previously described [35,36].
pension was achieved. The GL-1-containing liposomes and emulsions were A complex was formed between 10 ␮g of plasmid DNA and TransIT poly-
stored at 4⬚C until ready for use. Liposomal formulations containing a flu- mer which was then rapidly delivered (6–8 seconds) into the tail vein of a
orescent analog of GL-1, BODIPY FL C12-glucocerebroside (GL-1B; Molecular mouse. The volume administered (in ml) was equal to 1/10th the weight of
Probes, Eugene, OR) were prepared by the same methodology. The amount the mouse (in g).

MOLECULAR THERAPY Vol. 6, No. 2, August 2002 187


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL

Measurement of glucocerebrosidase activity. Tissues were homogenized antibody (BD Biosciences, San Diego, CA) or a sheep anti-rat von
in 150 mM citrate-phosphate buffer, pH 5.4, containing sodium tauro- Willebrand factor (endothelial cell marker) antibody (Accurate Chemical
cholate (0.25%, w/w) and Triton X-100 (0.25%, w/w) using a Tissue Tearor Co., Westbury, NY). Following three 5-minute washes with 1X Cadenza
(BioSpec Products Inc., Bartlesville, OK). The homogenates were clarified buffer (Shandon, Pittsburgh, PA), the slides were incubated for 1 hour with
by centrifugation for 15 minutes at 10000g and then assayed for gluco- the appropriate secondary antibodies. These were either a goat anti-mouse
cerebrosidase activity. The enzymatic activity of glucocerebrosidase was IgG2a AlexaFluor488, or a donkey anti-sheep IgG AlexaFluor488 (Molecular
determined using the synthetic substrate 4-methylumbelliferyl-glucopyra- Probes). Sections were viewed using an Olympus BX60 fluorescent micro-
noside (Sigma, St. Louis, MO) at 10 mM in homogenization buffer con- scope, and images were captured using Magnafire v1.0 (Lead Technologies)
taining 1% bovine serum albumin at 37⬚C. The reaction was terminated with an Optronics digital imager.
by adding 0.5 volume of 1M glycine buffer, pH 12.5, and the cleaved 4-
methylumbelliferone measured using a fluorimeter (Ex365/Em445). ACKNOWLEDGMENTS
Quantitation of tissue glucosylceramide levels. Tissue GL-1 levels were We thank Ronald Scheule (Genzyme Corporation) for his insights and review of
quantified using a modification of the procedures described [45–47]. Briefly, the manuscript and the Vector Production, AC&TS, and Histology groups for
total lipid from 100 mg portions of the test tissue was extracted by homog- their technical assistance.
enizing in 2 ml of a 2:1 (v/v) chloroform:methanol mixture using a
TissueTearor (BioSpec Products Inc., Bartlesville, OK). Following a 15- RECEIVED FOR PUBLICATION MARCH 18, 2002; ACCEPTED MAY 22, 2002.
minute incubation at 37⬚C, tissue debris was pelleted by centrifugation for
5 minutes at 1000g. The supernatant was removed and saved (sample 1)
REFERENCES
and the pellet was re-extracted with 2 ml of the 2:1 chloroform:methanol 1. Beutler, E., and Grabowski, G. A. (2001). Gaucher disease. In The Metabolic and Molecular
mix. This was again incubated for 15 minutes at 37⬚C, after which the Basis of Inherited Disease (C. R. Scriver, A. L. Beaudet, D. Valle, and W. S. Sly, Eds.), pp.
sample was re-centrifuged, and the supernatant was removed and combined 3635-3668. McGraw Hill, New York.
with sample 1. The combined supernatants were extracted with 0.22 vol- 2. Morales, L. E. (1996). Gauchers disease: a Review. Ann. Pharmacother. 30: 381–388.
umes of 0.9% saline by vortexing for 1 minute. The phases were separated 3. Grabowski, G. A., and Horowitz, M. (1997). Gauchers disease: molecular, genetic and
by centrifugation for 5 minutes at 1000g, which resulted in a lower organic enzymological aspects. Baill. Clin. Hæmatol. 10: 635–656.
4. Furbish, F. S., Steer, C. J., Krett, N. L., and Barranger, J. A. (1981). Uptake and distribu-
phase (sample 2) and an upper aqueous phase. The aqueous phase was
tion of placental glucocerebrosidase in rat hepatic cells and effects of sequential degly-
removed and re-extracted with 4.5 volumes of the 2:1 chloroform:methanol
cosylation. Biochim. Biophys. Acta 673: 425–434.
mixture. Following centrifugation, the aqueous phase was carefully 5. Ciana, G., Cuttini, M., and Bembi, B. (1997). Short-term effects of pamidronate in
removed and the organic phase was combined with sample 2. The com- patients with Gaucher’s disease and severe skeletal involvement. New Engl. J. Med. 337:
bined organic phases were re-extracted with 0.22 volumes of 0.9% saline, 712.
after which the final organic phase was dried down to a film. The film was 6. Lee, L., Abe, A., and Shayman, J. A. (1999). Improved inhibitors of glucosylceramide syn-
solubilized in 1 ml chloroform and a 200 ␮l aliquot passed over a LiChrolut thase. J. Biol. Chem. 274: 14662–14669.
RP-18 solid-phase extraction column (Merck, Darmstadt, Germany). After 7. Cox, T., et al. (2000). Novel oral treatment of Gauchers disease with N-butyldeoxyno-
jirimycin (OGT 918) to decrease substrate biosynthesis. Lancet 355: 1481–1485.
washing the column with 2 ml of chloroform, neutral glycolipids were
8. Liu, C., Dunigan, J. T., Watkins, S. C., Bahnson, A. B., and Barranger, J. A. (1998). Long-
eluted with 1 ml of acetone:methanol (9:1, v/v). The eluted samples were
term expression, systemic delivery, and macrophage uptake of recombinant human glu-
dried and solubilized in 200 ␮l of chloroform, 50 ␮l of which was then spot- cocerebrosidase in mice transplanted with genetically modified primary myoblasts. Hum.
ted onto an HPTLC plate (#13728 ; Merck, Darmstadt, Germany) along with Gene Ther. 9: 2375–2384.
known GL-1 standards (Matreya, Inc., Pleasant Gap, PA). The lipids were 9. Ross, C. J. D., Bastedo, L., Maier, S. A., Sands, M. S., and Chang, P. L. (2000). Treatment
chromatographed in a tank equilibrated with chloroform:methanol:water of a lysosomal storage disease, mucopolysaccharidosis VII, with microencapsulated
(70:30:10, v/v) for approximately 25 minutes. The plates were dried, recombinant cells. Hum. Gene Ther. 11: 2117–2127.
sprayed with 3% (w/v) cupric acetate monohydrate and 15% (v/v) phos- 10. Correll, P. H., Colilla, S., Dave, H. P. G., and Karlsson, S. (1992). High levels of human
glucocerebrosidase activity in macrophages of long-term reconstituted mice after retro-
phoric acid, and baked for 20–30 minutes at 180⬚C to develop the sepa-
viral infection of hematopoietic stem cells. Blood 80: 331–336.
rated lipid bands. The baked TLC plates were scanned as high-resolution
11. Xu, L. C., et al. (1995). Long-term in vivo expression of the human glucocerebrosidase
TIFF images, and the bands were quantified using ScanAnalysis (Biosoft, gene in nonhuman primates after CD34+ hematopoietic cell transduction with cell-free
Ferguson, MO). Statistical significance was determined using an unpaired retroviral vector preparations. Proc. Natl. Acad. Sci. USA 92: 4372–4376.
t-test (Statview; SAS Inst. Inc., Cary, NC). 12. Dunbar, C. E., et al. (1998). Retroviral transfer of the glucocerebrosidase gene into
CD34+ cells from patients with Gaucher disease: In vivo detection of transduced cells
Histology and immunofluorescence staining. Female Fischer 344/NTac
without myeloablation. Hum. Gene Ther. 9: 2629–2640.
rats (6–8 weeks old) were obtained from Taconic (Germantown, NY).
13. Havenga, M. J. E., Valerio, D., Hoogerbrugge, P., and Van Es, H. H. G. (1999). In vivo
Following treatment with either the recombinant enzymes or gene delivery methotrexate selection of murine hemopoietic cells transduced with a retroviral vector
vectors, the rats were anesthetized with an intraperitoneal injection of ket- for Gaucher disease. Gene Ther. 6: 1661–1669.
amine/xylamine. A whole body perfusion was performed by way of the left 14. Ohashi, T., et al. (1992). Efficient transfer and sustained high expression of the human
ventricle of the heart using a butterfly needle. PBS at 4⬚C containing 10 U/ml glucocerebrosidase gene in mice and their functional macrophages following trans-
heparin was perfused at a rate of 82 ml/minute until the lungs were plantation of bone marrow transduced by a retroviral vector. Proc. Natl. Acad. Sci. USA
blanched. The inferior vena cava was cut and the flow of PBS/heparin con- 89: 11332–11326.
15. Novelli, E.M., and Barranger, J.A. (2001). Gene therapy for lysosomal storage disorders.
tinued until the color of the liver changed from dark red to a light brown.
Expert Opin. Biol. Ther. 1: 857–867.
The rat was then perfused with approximately 200 ml/kg fixative (2% (v/v)
16. Tybulewicz, V. L. J., et al. (1992). Animal model of Gauchers disease from targeted dis-
formaldehyde, 0.0156% (v/v) glutaraldehyde in PBS) at 4⬚C. Following per- ruption of the mouse glucocerebrosidase Gene. Nature 357: 407–410.
fusion, the liver was excised, covered with fixative solution and incubated 17. Bornstein, P., et al. (1995). Metaxin, a gene contiguous to both thrombospondin 3 and
at room temperature for 1 hour. It was then immersed in a 30% sucrose solu- glucocerebrosidase, is required for embryonic development in the mouse: Implications
tion at 4⬚C for 24 hours before being embedded in Tissue-Tek OCT com- for Gaucher disease. Proc. Natl. Acad. Sci. USA 92: 4547–4551.
pound (Sakura Inc., Torrance, CA) and microtomed into 5 ␮m thick sections. 18. Liu, Y., et al. (1998). Mice with type 2 and 3 Gaucher disease point mutations gener-
All antibody incubations were performed in block solution (PBS con- ated by a single insertion mutagenesis procedure (SIMP). Proc. Natl. Acad. Sci. USA 95:
2503–2508.
taining 3% normal goat serum). To localize human glucocerebrosidase,
19. Sidransky, E., and Ginns, E. I. (1997). Gauchers disease: the best laid schemes of mice
liver sections were first incubated for 1 hour with an equal mixture of 1B5
and men. Baill. Clin. Hæmatol. 10: 725–737.
and 8E4 [48], two human specific anti-glucocerebrosidase monoclonal anti- 20. Holleran, W. M., et al. (1994). Consequences of ␤-glucocerebrosidase deficiency in epi-
bodies. Both of these antibodies are of the IgG1␬ isotype. To visualize the dermis: ultrastructure and permeability barrier alteration in Gaucher disease. J. Clin.
enzyme, the sections were treated with an AlexaFluor568-labeled goat anti- Invest. 91: 1736–1764.
mouse IgG1 secondary antibody. The liver sections were simultaneously 21. Kanfer, J. N., Legler, G., Sullivan, L., Raghavan, S., and Mumford, R. A. (1975). The
probed with either a mouse anti-rat CD11b (IgG2a; macrophage marker) Gaucher mouse. Biochem. Biophys. Res. Comm. 67: 85–90.

188 MOLECULAR THERAPY Vol. 6, No. 2, August 2002


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2002.0650, available online at http://www.idealibrary.com on IDEAL
ARTICLE

22. Stephens, M. C., Bernatsky, A., Burachinsky, V., Legler, G., and Kanfer, J. N. (1978). The 35. Zhang, G., Budker, V., and Wolff, J. A. (1999). High levels of foreign gene expression in
Gaucher mouse: differential action of conduritol B epoxide and reversibility of its effects. hepatocytes after tail vein injections of naked plasmid DNA. Hum. Gene Ther. 10:
J. Neurochem. 30: 1023–1027. 1735–1737.
23. Legler, G. (1990). Glycoside hydrolases: mechanistic information from studies with 36. Liu, F., Song, Y. K., and Liu, D. (1999). Hydrodynamics-based transfection in animals by
reversible and irreversible inhibitors. Adv. Carbohydr. Chem. Biochem. 48: 319–384. systemic administration of plasmid DNA. Gene Ther. 6: 1258–1266.
24. Hara, A., and Radin, N. S. (1979). Destruction and resynthesis of mouse ␤-glucosidases. 37. Li, C., et al. (2002). Adenovirus-transduced lung as a portal for delivering ␣-galactosi-
Biochim. Biophys. Acta 582: 412–422. dase A into systemic circulation for Fabry disease. Mol. Ther. 5: 745–754.
25. Datta, S. C., and Radin, N. S. (1988). Normalization of liver glucosylceramide levels in 38. Tokoro, T., Gal, A. E., Galo, L. L., and Brady, R. O. (1987). Studies of the pathogenesis
the Gaucher mouse by phosphatidylserine injection. Biochem. Biophys. Res. Comm. 152: of Gauchers disease: tissue distribution and biliary excretion of [14C]L-glucosylceramide
155–160. in rats. J. Lipid Res. 28: 968–972.
26. Pentchev, P. G., et al. (1981). Biliary excretion of glycolipid in induced or inherited glu- 39. Brady, R. O., Pentchev, P. G., Gal, A. E., Hibbert, S. R., and Dekaban, A. S. (1974).
cosylceramide lipidosis. Biochim. Biophys. Acta 665: 615–618. Replacement therapy for inherited enzyme deficiency. Use of purified glucocerebrosi-
27. Ellens, H., Morselt, H., and Scherpof, G. (1981). In vivo fate of large unilamellar sphin- dase in Gauchers disease. New Eng. J. Med. 291: 989–993.
gomyelin-cholesterol liposomes after intraperitoneal and intravenous injection into rats. 40. Morrone, S., Pentchev, P. G., Baynes, J., and Thorpe, S. (1981). Studies in vivo of the
Biochim. Biophys. Acta 674: 10–18. tissue uptake, cellular distribution and catabolic turnover of exogenous glucocerebrosi-
28. Worgall, S., Wolff, G., Falck-Pedersen, E., Crystal, R.G. (1997). Innate immune mecha- dase in rat. Biochem. J. 194: 733–742.
nisms dominate elimination of adenoviral vectors following in vivo administration. Hum. 41. Doebber, T. W., et al. (1982). Enhanced macrophage uptake of synthetically glycosy-
Gene Ther. 8: 37–44. lated human placental ␤-glucocerebrosidase. J. Biol. Chem. 257: 2193–2199.
29. van Rooijen, N. and Sanders, A. (1994). Liposome mediated depletion of macrophages: 42. Liu, C., Duniga, J. T., Watkins, S. C., Bahnson, A. B., and Barranger, J. A. (1998). Long-
mechanism of action, preparation of liposomes and applications. J. Immunol. Methods term expression, systemic delivery, and macrophage uptake of recombinant human glu-
174: 83–93. cocerebrosidase in mice transplanted with genetically modified primary myoblasts. Hum.
30. van Rooijen, N., Sanders, A., and van der Berg, T. K. (1996). Apoptosis of macrophages Gene Ther. 9: 2375–2384.
induced by liposome-mediated intracellular delivery of clodronate and propamidine. J. 43. Armentano, D., et al. (1997). Effect of the E4 region on the persistence of transgene
Immunol. Methods 193: 93–99. expression from adenovirus vectors. J. Virol. 71: 2408–2416.
31. Barton, N.W., Furbish, F.S., Murray, G.J., Garfield, M., and Brady, R.O. (1990). 44. Yew, N. S., et al. (2000). Reduced inflammatory response to plasmid DNA vectors by
Therapeutic response to intravenous infusions of glucocerebrosidase in a patient with elimination and inhibition of immunostimulatory CpG motifs. Mol. Ther. 1: 255–262,
Gaucher disease. Proc. Natl. Acad. Sci USA 87: 1913–1990. doi:10.1006/mthe.2002.0036.
32. Xu, L. C., et al. (1996). Turnover and distribution of intravenously administered man- 45. Miller, S. P. F., Zirzow, G. C., Doppelt, S. H., Brady, R. O., and Barton, N. W. (1996).
nose-terminated human acid ␤-glucosidase in murine and human tissues. Pediatr. Res. Analysis of the lipids of normal and Gaucher bone marrow. J. Lab. Clin. Med. 127:
39: 313–322. 353–358.
33. Kuzmin, A. I., Finegold, M. J., and Eisensmith, R. C. (1997). Macrophage depletion 46. Doering, T., et al. (1999). Sphingolipid activator proteins are required for epidermal per-
increases the safety, efficacy and persistence of adenovirus-mediated gene transfer in vivo. meability barrier formation. J. Biol. Chem. 274: 11038–11045.
Gene Ther. 4: 309–316. 47. Abe, A., et al. (2000). Glycosphingolipid depletion in Fabry disease lymphoblasts with
34. Wolff, G., et al. (1997). Enhancement of in vivo adenovirus-mediated gene transfer and potent inhibitors of glucosylceramide synthase. Kidney Int. 57: 446–454.
expression by prior depletion of tissue macrophages in the target organ. J. Virol. 71: 48. Aerts, J. M. F. G., et al. (1990). Comparative study on glucocerebrosidase in spleens from
624–629. patients with Gaucher disease. Biochem. J. 269: 93–100.

MOLECULAR THERAPY Vol. 6, No. 2, August 2002 189


Copyright © The American Society of Gene Therapy

You might also like