Accepted Manuscript Adeno-associated viral gene therapy corrects a mouse model of argininosuccinic aciduria
Scott N. Ashley, Jayme M.L. Nordin, Jenny A. Greig, James M. Wilson PII: DOI: Reference:
S1096-7192(18)30195-1 doi:10.1016/j.ymgme.2018.08.013 YMGME 6399
To appear in:
Molecular Genetics and Metabolism
Received date: Revised date: Accepted date:
9 April 2018 26 August 2018 27 August 2018
Please cite this article as: Scott N. Ashley, Jayme M.L. Nordin, Jenny A. Greig, James M. Wilson , Adeno-associated viral gene therapy corrects a mouse model of argininosuccinic aciduria. Ymgme (2018), doi:10.1016/j.ymgme.2018.08.013
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Adeno-Associated Viral Gene Therapy Corrects a Mouse Model of Argininosuccinic Aciduria Scott N. Ashley, Jayme M. L. Nordin, Jenny A. Greig, and James M. Wilson*
T
Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA,
CR
IP
USA
*Correspondence should be addressed to J.M.W.:
US
James M. Wilson, M.D., Ph.D.
AN
Gene Therapy Program Perelman School of Medicine
M
University of Pennsylvania
Philadelphia, PA 19104, USA
ED
125 South 31st Street
PT
Phone: 215-898-0920; Fax: 215-494-5444
AC
CE
E-mail:
[email protected]
ACCEPTED MANUSCRIPT Abstract Argininosuccinic aciduria (ASA) is the second most common genetic disorder affecting the urea cycle. The disease is caused by deleterious mutations in the gene encoding argininosuccinate lyase (ASL); total loss of ASL activity results in severe neonatal onset of the disease, which is
The long-term complications of ASA, such as hypertension and neurocognitive
IP
and death.
T
characterized by hyperammonemia within a few days of birth that can rapidly progress to coma
CR
deficits, appear to be resistant to the current treatment options of dietary restriction, arginine supplementation, and nitrogen scavenging drugs. Treatment-resistant disease is currently being
US
managed by orthotopic liver transplant, which shows variable improvement and requires lifetime
AN
immunosuppression. Here, we developed a gene therapy strategy for ASA aimed at alleviating the symptoms associated with urea cycle disruption by providing stable expression of ASL We designed a codon-optimized human ASL gene packaged within an
M
protein in the liver.
ED
adeno-associated virus serotype 8 (AAV8) as a vector for targeted delivery to the liver. To evaluate the therapeutic efficacy of this approach, we utilized a murine hypomorphic model of
PT
ASA. Neonatal administration of AAV8 via the temporal facial vein extended survival in ASA
CE
hypomorphic mice, although not to wild-type levels.
Intravenous injection into adolescent
hypomorphic mice led to increased survival and body weight and correction of metabolites
AC
associated with the disease.
Our results demonstrate that AAV8 gene therapy is a viable
approach for the treatment of ASA.
Highlights:
AAV gene therapy extends survival of adolescent ASA hypomorphic mice
AAV gene therapy extends survival of neonatal ASA hypomorphic mice
2
ACCEPTED MANUSCRIPT
AAV gene therapy normalizes plasma argininosuccinic acid and citrulline levels in ASA hypomorphic mice
AAV gene therapy normalizes serum transaminase levels in ASA hypomorphic mice
T
Keywords: AAV, Gene Therapy, Argininosuccinic Aciduria, Liver, Urea Cycle
IP
Abbreviations: Argininosuccinic aciduria (ASA); argininosuccinate lyase (ASL); adeno-
CR
associated virus (AAV); thyroxine-binding globulin (TBG); bovine growth hormone (bGH);
US
genome copies (GC); intravenous (IV); analysis of variance (ANOVA); immunohistochemistry
AC
CE
PT
ED
M
AN
(IHC); alanine aminotransferase (ALT); aspartate aminotransferase (AST)
3
ACCEPTED MANUSCRIPT 1. Introduction Argininosuccinic aciduria (ASA) is an autosomal recessive disorder of the urea cycle caused by mutations within the argininosuccinate lyase (ASL) gene that result in impaired arginine synthesis. Newborn screening programs in the United States are important for early detection of
diagnosis
is
determined by detection of the unique metabolite marker
IP
differential
T
ureagenesis disorders; however, due to the similarity of ASA to other urea cycle disorders,
Neonatal
ASA, characterized by
US
primary manifestations: neonatal and late onset.
CR
argininosuccinic acid by tandem mass spectrometry in patient plasma [1, 2]. The disease has two
hyperammonemia within days following birth, can be treated by hemodialysis followed by
AN
lifetime maintenance care to reduce the risk of further episodes [3-5]. Late-onset ASA has a less
M
severe phenotype that includes episodic hyperammonemia triggered by acute infection or stress , as well as neurocognitive impairment with associated learning or behavioral abnormalities [4].
ED
Patients with ASA can also manifest symptoms unrelated to hyperammonemia due to the lack of
PT
nitric oxide production in both forms of the disorder, including neurocognitive deficiencies, cirrhosis of the liver, and systemic hypertension [4-7].
Standard maintenance care includes
CE
protein restriction, arginine supplementation, and nitrogen scavenging drugs (e.g., sodium
AC
benzoate, sodium phenylburyrate, and glycerolphenylburyrate) [8, 9]. Individuals with recurrent hyperammonemia or cirrhosis of the liver can also undergo liver transplant as a diseasemodifying process. However, lifetime immunosuppression is required following transplant and early transplantation cases showed lower levels of improvement compared to other urea cycle disorders, although more recent case studies showed positive health and quality of life outcomes [10-14].
4
ACCEPTED MANUSCRIPT As the absence of ASL from hepatocytes causes ASA-related hyperammonemia [12, 15, 16], liver-targeted gene therapy has the potential to benefit patients with severe disease. Adenoassociated virus (AAV)-mediated gene therapy has demonstrated both therapeutic efficacy and low immunogenicity in murine models for the treatment of other urea cycle disorders (e.g.,
T
ornithine transcarbamylase deficiency, arginase deficiency, and citrullinemia) [17-23].
IP
However, ASA presents a unique challenge as the enzyme is essential not only for the removal
CR
of waste nitrogen via the urea cycle, but also for the clearance of argininosuccinic acid through its conversion to arginine. The buildup of argininosuccinic acid is thought to cause some of the
US
unique symptoms of the disease, including hepatic and cognitive deficits [4, 5]. Based on results
AN
from patients with ASA who have received liver transplants [15, 16, 24], liver-targeted gene therapy is uniquely positioned to restore the urea cycle and increase quality of life without the
M
need for a highly invasive procedure or a continued drug regimen. In this study, we developed a
ED
gene therapy for ASA using an AAV8 vector expressing the human ASL gene under the control of the liver-specific thyroxine-binding globulin (TBG) promoter, and examined therapeutic
AC
CE
PT
efficacy in both neonatal and adolescent ASA hypomorphic mice.
5
ACCEPTED MANUSCRIPT 2. Materials and Methods 2.1. Mice ASA hypomorphic mice on a C57BL/6 background were acquired from The Jackson Laboratory (Bar Harbor, ME) and a colony was maintained at the University of Pennsylvania under specific All animal procedures and protocols were approved by the
T
pathogen-free conditions.
CR
IP
Institutional Animal Care and Use Committee of the University of Pennsylvania.
2.2. Vectors
promoter
with
a
(AAV8.TBG.hASLco.bGH).
bovine
growth
hormone
(bGH)
polyA
signal
sequence
AN
TBG
US
AAV8 vectors expressing human codon-optimized ASL were designed under the control of a
AAV vectors were produced by the Penn Vector Core at the
2.3. Neonatal gene therapy
ED
M
University of Pennsylvania, as previously described [25].
PT
During the first 24 h following birth, ASA hypomorphic pups (n=4 low dose, n=7 high dose)
CE
received intravenous (IV) administration of 1010 or 1011 genome copies (GC)/mouse of AAV8.TBG.hASLco.bGH via the temporal facial vein.
Mice were weighed throughout the
AC
study and genotyped upon weaning. Mice were sacrificed upon reaching median survival, and tissues were collected for histology.
2.4. Adolescent gene therapy ASA hypomorphic mice four to five weeks of age received IV administration of 1013 or 6x1013 GC/kg of AAV8.TBG.hASLco.bGH via the retro orbital vein (n=8).
6
Mice were weighed
ACCEPTED MANUSCRIPT throughout the study.
Sub-mandibular bleeds were performed weekly to collect plasma for
monitoring liver transaminases and urea cycle metabolites.
2.5. Serum analyses
T
Plasma samples were submitted to Antech Diagnostics (Irvine, CA) for analysis of liver
IP
transaminases and Charles River Laboratories (Worcester, MA) for analysis of arginine,
acetonitrile.
CR
citrulline, and argininosuccinic acid by LC-MS/MS following protein precipitation with Mice were sacrificed after three months and tissues were collected for
AN
US
biodistribution and histology.
2.6. ASL immunohistochemistry
M
Tissues were fixed in formalin for a minimum of 24 h and then paraffin embedded. Sections
ED
were deparaffinized through an ethanol and xylene series, boiled for 6 min in 10 mM citrate buffer (pH 6.0) for antigen retrieval, and sequentially treated with 2% H 2O2 (15 min),
PT
avidin/biotin blocking reagents (15 min each; Vector Laboratories, Burlingame, CA), and Sections were then
CE
blocking buffer (10 min; 1% donkey serum in PBS + 0.2% Triton).
incubated with a rabbit serum against ASL (1 h; HPA016646, Sigma-Aldrich, St. Louis, MO)
AC
and biotinylated secondary anti-rabbit antibodies (45 min; Jackson ImmunoResearch, Westgrove, PA) diluted in blocking buffer at the manufacturer’s recommended concentration. A Vectastain Elite ABC Kit (Vector Laboratories, Burlingame, CA) was used according to the manufacturer's instructions with 3,3'-diaminobenzidine as the substrate to stain bound antibodies.
2.7. Sirius Red and HE
7
ACCEPTED MANUSCRIPT To detect liver fibrosis, Sirius red staining was performed on paraffin sections. Deparaffinized sections were stained for 90 min in a solution containing 0.1% (w/v) Direct Red (Sigma) and 4% (w/v) picric acid (Sigma-Aldrich, St. Louis, MO), and then washed with 0.01 N HCl (2 x 1 min), dehydrated through an ethanol and xylene series, and coverslipped. Paraffin sections were
IP
T
stained with hematoxylin and eosin (H&E) according to standard protocols.
CR
2.8. Biodistribution
Liver samples were frozen on dry ice at the time of necropsy, and DNA was extracted using the
US
QIAamp DNA Mini Kit (Qiagen, Valencia, CA). Detection and quantification of vector GCs in
AN
extracted DNA were performed by real-time polymerase chain reaction, as previously described [26]. Briefly, genomic DNA was isolated, and vector GCs were quantified using primers/probes
M
designed against the promoter sequence of the vector. Quantification of GCs from liver was
PT
2.9. ASL activity assay
ED
performed on one liver sample from each mouse.
CE
Liver (25-30 mg) was added to 200 µl of cold homogenizing buffer containing 50 mM phosphate buffer (pH 7.5) and proteinase inhibitors (EDTA-free proteinase inhibitor cocktail; Roche, Basel,
AC
Switzerland), and homogenized using Tissue Lyser II (Qiagen, Valencia, CA), at a frequency of 30 for 30 seconds.
Homogenates were centrifuged at 10,000 x g for 20 min at 4ºC, and
supernatants were kept frozen at 80ºC. Lysate (2 µl) was added to 48 µl of 50 mM phosphate buffer (pH 7.3), 3.6 mM argininosuccinic acid (Sigma Aldrich, St. Louis, MO). The reaction was incubated at 37ºC for 1 h and stopped by heating at 80ºC for 20 min. Fumarate was measured with Fumarate Assay Kit (Sigma Aldrich, St. Louis, MO) per the manufacturer’s
8
ACCEPTED MANUSCRIPT specifications using 5 µl of reaction sample mixture.
2.10. Statistical analysis Comparison of survival was performed by logrank test with each experimental group compared
T
to uninjected control groups, unless otherwise stated. Other parameters were compared by one-
IP
way analysis of variance (ANOVA) with Dunnett’s multiple comparison test comparing each
CR
group to the untreated wild-type control group, unless otherwise stated. *p < 0.05, **p < 0.01,
US
***p < 0.001.
AN
3. Results
3.1. An ASA hypomorphic mouse model enables the study of both neonatal and adolescent mice
M
To evaluate our gene therapy approach, we first considered which of the two ASA mouse models
ED
to use for our studies. A knockout model (Asl -/-) was created by replacing exons 8 and 9 with a 1,400 bp neomycin cassette, resulting in a frame shift in the mRNA beginning with exon 10 [27].
PT
All Asl-/- mice have elevated plasma ammonia, argininosuccinic acid, and citrulline, as well as
CE
low plasma arginine. However, these mice die within 48 hours of birth, so this model can only be used to study treatment of a neonatal cohort. As an alternative, an ASA hypomorphic mouse
AC
model has been developed, where a 1,200 bp neomycin cassette was inserted into intron 9, resulting in reduced, but not ablated, mRNA levels and slightly prolonged survival [6]. These mice have the same characteristic variations in amino acids and liver metabolites, and they display a sparse fur coat. Therefore, for the current study we used the ASA hypomorphic mouse. We confirmed that these mice have a mean survival of 22 days (Figure 1A). Homozygous ASA hypomorphic pups were indistinguishable from heterozygous or wild-type littermates until five
9
ACCEPTED MANUSCRIPT days after birth, at which time the hypomorphic mice failed to develop a fur coat and were visibly smaller.
3.2. Prophylactic AAV8 gene therapy extends survival of neonatal ASA hypomorphic mice
T
We next evaluated AAV8 therapy in neonatal mice. We performed IV injections via the temporal
We genotyped mice after weaning and monitored the vector-administered
CR
24 hours of birth.
IP
facial vein with 1010 or 1011 GC/mouse of the AAV8.TBG.hASLco.bGH vector within the first
ASA hypomorphic mice for survival. Vector administration at both doses significantly increased
US
survival compared to untreated ASA hypomorphic mice (p < 0.01; Figure 1B). The increase in
AN
median survival was dose dependent, with a median survival of 165 days for the 1011 GC/mouse group compared to 136 days for the 1010 GC/mouse group. Weight gain for vector-administered
M
ASA hypomorphic mice was comparable to wild-type littermates throughout the study, with the
ED
exception of the female vector-administered hypomorphic mice at the last time point evaluated
PT
(day 63; Figures 1C).
We used
CE
Once the mice reached median survival, we euthanized and necropsied them.
immunohistochemistry (IHC) of the liver to visualize ASL protein expression (Figure 2). IHC
AC
revealed a small number of strongly stained positive cells. The low level of transgene expression observed in these adolescent mice was expected, as the rapid proliferation of hepatocytes during early development has been shown to significantly dilute non-integrating vector genomes [2830].
Despite the low level of transgene expression observed at the time of necropsy, ASL
activity was sufficient to allow all vector-treated ASA hypomorphic mice to live beyond the 22 day mean survival of untreated ASA hypomorphic mice. However, as expected without re-
10
ACCEPTED MANUSCRIPT administration, we confirmed that vector dilution as the animal grew led to reduced efficacy and resulted in premature death (Figure 1B).
3.3. AAV8 gene therapy treatment extends survival, increases weight, and normalizes serum
T
transaminase levels in adolescent ASA hypomorphic mice
IP
In addition to a prophylactic gene therapy model following neonatal administration, we
CR
investigated the potential treatment effects of AAV8 gene therapy in adolescent ASA hypomorphic mice. We administered AAV8 to 30-day-old adolescent mice via the retro orbital
US
vein; the standard adolescent mouse IV administration route via the tail vein could not be used
AN
for these adolescent ASA hypomorphic mice as their average weight was 8.8 g. We collected plasma from the first cohort of female mice for baseline metabolite and transaminase evaluation
M
prior to IV administration with 6x1013 GC/kg of vector.
Mice that had been bled had high
ED
mortality rates prior to or shortly after vector administration, likely due to reduced blood volume and were omitted from the survival data set for this cohort (Figure 3A). Therefore, we did not
PT
determine baseline values for the other cohorts. Further cohorts of adolescent ASA hypomorphic
CE
mice were either untreated or administered IV with 1013 GC/kg of vector. Survival of mice in all vector-administered cohorts increased compared to untreated ASA hypomorphic mice (p <
AC
0.001; Figure 3A). Mean survival was extended to 91 days in the low-dose female group, with the study terminated before the high-dose female and low-dose male cohorts had lost enough mice to determine mean survival (Figure 3A). We observed a sex difference in survival, with untreated female ASA hypomorphic mice having increased mean survival compared to untreated male ASA hypomorphic mice (p < 0.001). However, following low-dose vector administration, male mice showed increased survival and weight gain compared to female mice, which could
11
ACCEPTED MANUSCRIPT possibly be due to an androgen-dependent, species-specific effect on AAV transduction of the liver as previously described [31]. We observed a dose-dependent increase in weight gain; however, even mice administered 6x1013 GC/kg of vector did not match wild-type body weights (Figure 3B). All vector-treated groups had reductions in plasma alanine aminotransferase (ALT)
T
and aspartate aminotransferase (AST) compared to untreated controls (Figure 3C and 3D).
IP
3.4. AAV gene therapy normalizes plasma argininosuccinic acid and citrulline levels in ASA
CR
hypomorphic mice
AAV gene therapy has the unique potential to restore the urea cycle in hepatocytes without the Therefore, we evaluated the effect of our AAV8 vector on
US
need for a liver transplant.
AN
metabolites associated with ASA and other aspects of the urea cycle. Argininosuccinic acid is broken down into arginine and fumarate by the ASL enzyme, leading to elevations in
M
argininosuccinic acid and citrulline (a substrate of the reaction upstream of ASL), and reductions
ED
in arginine.
PT
Plasma citrulline was elevated to 474 ± 155 µmol in the hypomorphic mice compared to 58.3 ± In the high-dose female and low-dose male
CE
14.4 µmol in the wild-type mice (Figure 4A).
groups, we saw a dramatic reduction of citrulline in plasma, resulting in no significant difference
AC
in citrulline levels compared to wild-type littermates indicating correction of the metabolite (Figure 4A). We did not observe an effect of the low dose of vector on plasma citrulline levels in the female mice. Plasma argininosuccinic acid, which is the metabolite that differentiates ASA from other urea cycle disorders and is thought to play a role in the unique symptoms of the disease, was elevated from 163.6 ± 60.0 µmol in the hypomorphic mice compared to 1.53 ± 1.13 µmol in the wild-type mice (Figure 4B). Plasma argininosuccinic acid was corrected to normal
12
ACCEPTED MANUSCRIPT levels in the high-dose female group (Figure 4B), but remained elevated in both the male and female low-dose cohorts, indicating a steep dose effect.
Wild-type mice had plasma arginine levels in the range of 45-185 µM with a mean of 95 µM Characterization of arginine levels in the ASA hypomorphic mouse was
T
(Figure 4C).
IP
complicated because of less dramatic differences between values in normal and ASA
CR
hypomorphic mice, and the early mortality of the mice in the absence of treatment. In fact, at the initiation of the study ASA hypomorphic mice had arginine levels similar to the wild type (75
US
µM) (Figure 4C). However, at day 35 we observed significant reductions in arginine levels from
AN
wild-type mice compared to male and female hypomorphic cohorts treated with 1013 GC/kg of AAV8 vector, suggesting incomplete correction. However, the extent of the therapeutic affect is
M
unclear as we do not have samples from untreated hypomorphic mice at this age group to analyze
ED
(Figure 4C). Incomplete correction may also be due to the liver-restricted nature of our therapy,
CE
synthesis in the body.
PT
where to correct arginine we would want to target to the kidney, the primary site of arginine
Upon termination of the study, we sacrificed the mice and harvested liver for determination of
AC
ASL protein distribution, liver disease progression, and vector GCs. Female ASA hypomorphic mice administered with the high dose of vector had a strong presence of ASL protein in the liver as measured by IHC (Figure 5A). Male and female mice dosed with the low dose showed similar levels of staining.
Activity of ASL protein in liver lysate of high-dose females as
measured by an activity assay was on average 25% of wild-type levels and statistically significantly higher than untreated hypomorphic mice, which had a mean activity of 3% (Figure
13
ACCEPTED MANUSCRIPT 5B). The activity in mice administered with the low vector dose was not statistically higher than in the untreated hypomorphic mice.
Vector genome copy analysis on liver samples demonstrated that ~four-fold higher vector GCs
T
were present in hepatocytes in the high-dose cohort compared to the low-dose groups (Figure
IP
5C). These results demonstrate that with sufficient transduction of liver by an ASL gene therapy
CR
vector, survival in the ASA hypomorphic mouse model can be greatly extended.
This is
AN
correction of metabolite markers to normal levels.
US
concurrent with a reduction in liver transaminases indicating reduced hepatic toxicity and the
The primary histopathologic findings in the liver of ASA hypomorph mice that received either
M
low- or high-dose vector included variable hepatocellular changes consisting of enlargement of
ED
both the nucleus and cytoplasm (karyocytomegaly) with and without individual hepatocellular necrosis and regeneration (indicated by increased incidence of mitotic figures). The more severe
PT
cases exhibited hepatocellular dissociation with and without lobular collapse. Other findings
CE
included mixed cellular infiltrates, fibrosis, and higher scores of bile duct and oval cell hyperplasia indicating a more severe phenotype (Figure 6). H&E staining revealed that low-dose
AC
male mice (Figure 7A and 8A) appeared to have more severe histopathological events than the equivalently dosed female cohort (Figure 7B); by contrast, the high-dose female group trended better than their low-dose counterpart (Figure 7C and 7D).
We observed minimal
histopathologic findings in untreated ASA hypomorphic mice compared to dosed cohorts (Figure 7E). We believe this result is due to the early death of these untreated mice (~ study day 15), as all of the histology of dosed groups were generated from samples taken at the termination of the
14
ACCEPTED MANUSCRIPT study (study day 98). In addition, we independently scored the liver fibrosis based on location and amount of hepatic infiltration on sections using Sirius red. Although we observed a range of scores for both doses, animals from the low-dose male cohort group (Figure 8B) more commonly showed the presence of and higher scores for fibrosis, compared to the low-dose (Figure 8C) or Again, untreated mice did not develop observable
CR
IP
histopathology due to the limited time for it to develop (Figure 8E).
T
high-dose (Figure 8D) female cohorts.
4. Discussion
US
ASA, caused by a deficiency in ASL activity, is characterized by the dysregulation of the urea
AN
cycle that inhibits arginine synthesis and nitric oxide production [4]. Here, we showed that delivery of the ASL gene into hepatocytes increased survival and weight gain of ASA
M
hypomorphic mice. Median survival for ASA hypomorphic mice was 22 days post birth due to a
ED
failure to thrive marked by an increase in liver transaminases and elevations in urea cycle metabolites. We were able to increase survival of these mice in a dose-dependent manner via
PT
neonatal administration of vector through the facial vein. However, treated mice did not gain
CE
weight to an equivalent degree as their wild-type littermates; liver histology suggested that dilution of vector genomes occurred with growth of the animal, which obscures regional
AC
transduction differences, consistent with what has been previously reported [28-30]. We believe that this vector dilution contributed to the early death of treated hypomorphic mice, where ASL expression necessary for survival may have dropped lower than a critical threshold resulting in a buildup of toxic metabolites. This will be a critically important phenomenon in advancing gene therapy for infant patients with ASA, as readministration of gene therapy may be necessary. Successful readministration of vector to the liver can prove difficult due to circulating
15
ACCEPTED MANUSCRIPT neutralizing antibodies to the vector capsid that would develop after the initial administration. However, recent advances in immune suppression have shown potential for either preventing AAV antibody development or reducing the circulating antibodies to other AAV serotypes,
T
allowing for repeat dosing [32, 33].
IP
To determine the efficacy of ASA gene therapy in more developed livers, we administered
CR
vector via a retro orbital IV injection to adolescent mice. These mice exhibited both increased survival and weight gain, although only the high dose resulted in survival similar to wild-type
US
littermates. One potential cause of the continued early morbidity in treated adolescent mice may
AN
be the vector genome dilution, because although these mice are older than the newborn treatment groups, they have not yet reached full size. Indeed, based on our monitoring of weight of
M
untreated ASA hypomorphic mice, a successful start to the therapy may ultimately result in an
ED
attenuation of its efficacy as it allows the treated mice to increase in weight rapidly.
PT
Cirrhosis of the liver is one of the hallmarks of ASA, affecting individuals more commonly than
CE
other urea cycle disorders, and is a primary reason that patients undergo liver transplant [3]. We did not observe liver pathology in untreated ASA hypomorphic mice following review of liver
AC
sections by a board-certified veterinary pathologist potentially because the affected mice died before cirrhosis could develop. However, we did observe extensive histopathology in some treated groups, especially the low-dose male cohort.
The low-dose female cohort had a
comparatively less severe phenotype that was further reduced in the high-dose cohort. These results could be indicative of a sex bias in treatment efficacy toward more correction in female mice; however, based on metabolite analysis of citrulline we might predict that the opposite is
16
ACCEPTED MANUSCRIPT occurring. Another possibility is that ASA liver pathology is more severe in male mice. This line of inquiry requires further understanding of the natural history of the ASA hypomorph, as no reports in the literature suggest a sex difference in the severity of ASA liver cirrhosis. Overall, the severity of histopathological findings in the female cohorts did seem to improve in a dose-
T
dependent manner indicating that gene therapy could be effective in treating one of the primary
IP
reasons patients with ASA require liver transplant [3]. We also observed reduced transaminase
CR
levels in all treated mouse groups at most time points studied. Furthermore, mice in the highdose group showed a trend for corrected arginine levels, indicating that AAV gene therapy might
US
correct arginine deficiency, which is not corrected sufficiently with liver transplant [3, 4]. The
AN
high-dose group also achieved normalization of plasma argininosuccinic acid. This encouraging
M
result suggests that argininosuccinic acid is being cleared from the liver.
ED
Citrulline, a metabolite upstream of ASL, was normalized in both high-dose administered females and low-dose administered males.
Evidence suggests that an androgen-dependent
PT
mechanism enhances transduction in male mice (27). However, we did not observe a difference
CE
in the vector genome copy number between low-dose groups based on sex, suggesting a potential
AC
difference in disease severity based on sex [31].
One of the major difficulties facing gene therapy treatment of ASA is the importance of ASL in non-hepatic metabolic pathways outside of the urea cycle and how they may negatively impact therapeutic efficacy [13, 14]. ASL is required for both cleavage of argininosuccinic acid into arginine in the kidneys, providing a major of source of serum arginine [34], and production of nitric oxide by endothelial cells lining the arteries [35]. Helper-dependent adenoviral vector
17
ACCEPTED MANUSCRIPT gene therapy for this hypomorphic mouse model of ASA showed similar improvements in repair of the urea cycle compared to our vector, but was unable to overcome other disease phenotypes associated with non-liver expression of ASL; we believe this would not be different for our gene therapy approach [36]. Initial longitudinal studies of orthotopic liver transplants for patients with
T
ASA, whose outcome a liver-targeted gene therapy could mimic, displayed a less positive
IP
outcome compared to other urea cycle disorders [14]. A debate has existed among researchers as
CR
to whether this low efficacy is due to insufficient arginine, loss of nitric oxide synthesis contributing to the cirrhosis of the liver, or buildup of argininosuccinic acid [12, 37]. However,
US
more recent studies of liver transplants have shown stronger positive outcomes [12, 15, 16].
AN
Taken together, it would seem prudent to consider continuing arginine supplementation after gene therapy, and with early treatment and improved care outcomes it may come to resemble
ED
M
more recent transplant success.
5. Conclusions
PT
In summary, we have demonstrated the feasibility of gene therapy for ASA using AAV8 vector
CE
technology. However, both routes of administration (IV via the temporal facial vein in newborns and retro orbitally in adolescent mice) pose some challenges. Neonatal administration was able
AC
to extend survival compared to untreated ASA hypomorphic mice, but not to the full lifespan of wild-type mice. This could be solved with readministration of the gene therapy vector at a later time point, an important consideration for future development.
Adolescent vector
administration, while able to correct metabolites at the highest dose tested, was less efficacious at a lower dose.
18
ACCEPTED MANUSCRIPT Acknowledgments We thank Deirdre McMenamin, Christine Draper, Hongwei Yu, and Meardey So for invaluable technical assistance.
T
Disclosures
IP
J.M. Wilson is an advisor to, holds equity in, and has a sponsored research agreement with
CR
REGENXBIO and Scout Bio; he also has a sponsored research agreement with Ultragenyx, Biogen, and Janssen, which are licensees of Penn technology. In addition, he has sponsored
US
research agreements with Precision Biosciences and Moderna Therapeutics.
J.M.W. is an
M
AN
inventor on patents that have been licensed to various biopharmaceutical companies.
ED
Funding Source
AC
CE
PT
This study was funded by internal funding from the University of Pennsylvania.
19
ACCEPTED MANUSCRIPT References [1]
R.D. Ganetzky, E. Bedoukian, M.A. Deardorff, C. Ficicioglu, Argininosuccinic Acid
Lyase Deficiency Missed by Newborn Screen JIMD reports (2016). [2]
J. Haberle, N. Boddaert, A. Burlina, A. Chakrapani, M. Dixon, M. Huemer, D. Karall, D.
T
Martinelli, P.S. Crespo, R. Santer, A. Servais, V. Valayannopoulos, M. Lindner, V. Rubio, C.
IP
Dionisi-Vici, Suggested guidelines for the diagnosis and management of urea cycle disorders
[3]
CR
Orphanet journal of rare diseases 7 (2012) 32.
S.C.S. Nagamani, A. Erez, B. Lee, Argininosuccinate Lyase Deficiency, in: R.A. Pagon,
US
M.P. Adam, H.H. Ardinger, S.E. Wallace, A. Amemiya, L.J.H. Bean, T.D. Bird, N. Ledbetter,
AN
H.C. Mefford, R.J.H. Smith, K. Stephens (Eds.), GeneReviews(R), University of Washington, Seattle
M
University of Washington, Seattle. GeneReviews is a registered trademark of the University of
A. Erez, Argininosuccinic aciduria: from a monogenic to a complex disorder Genet Med
15 (2013) 251-257.
H.A. Brusilow S, Urea cycle enzymes in: B.A. Scriver CR, Sly WS, Valle D, Childs B,
CE
[5]
PT
[4]
ED
Washington, Seattle. All rights reserved., Seattle (WA), 1993.
Kinzler KW, Vogelstein B (Ed.), The Metabolic and Molecular Bases of Inherited Disease,
[6]
AC
McGraw-Hill, New York, 2001. A. Erez, S.C. Nagamani, O.A. Shchelochkov, M.H. Premkumar, P.M. Campeau, Y.
Chen, H.K. Garg, L. Li, A. Mian, T.K. Bertin, J.O. Black, H. Zeng, Y. Tang, A.K. Reddy, M. Summar, W.E. O'Brien, D.G. Harrison, W.E. Mitch, J.C. Marini, J.L. Aschner, N.S. Bryan, B. Lee, Requirement of argininosuccinate lyase for systemic nitric oxide production Nat Med 17 (2011) 1619-1626.
20
ACCEPTED MANUSCRIPT [7]
M. Hermann, A. Flammer, T.F. Luscher, Nitric oxide in hypertension Journal of clinical
hypertension (Greenwich, Conn.) 8 (2006) 17-29. [8]
S.A. Berry, N. Longo, G.A. Diaz, S.E. McCandless, W.E. Smith, C.O. Harding, R. Zori,
C. Ficicioglu, U. Lichter-Konecki, B. Robinson, J. Vockley, Safety and efficacy of glycerol
T
phenylbutyrate for management of urea cycle disorders in patients aged 2months to 2years
J. Baruteau, E. Jameson, A.A. Morris, A. Chakrapani, S. Santra, S. Vijay, H. Kocadag,
CR
[9]
IP
Molecular genetics and metabolism 122 (2017) 46-53.
C.E. Beesley, S. Grunewald, E. Murphy, M. Cleary, H. Mundy, L. Abulhoul, A. Broomfield, R.
US
Lachmann, Y. Rahman, P.H. Robinson, L. MacPherson, K. Foster, W.K. Chong, D.A. Ridout,
AN
K.M. Bounford, S.N. Waddington, P.B. Mills, P. Gissen, J.E. Davison, Expanding the phenotype in argininosuccinic aciduria: need for new therapies Journal of inherited metabolic disease 40
M.L. Batshaw, R.B. MacArthur, M. Tuchman, Alternative pathway therapy for urea
ED
[10]
M
(2017) 357-368.
cycle disorders: twenty years later The Journal of pediatrics 138 (2001) S46-54; discussion S54-
C. Ficicioglu, R. Mandell, V.E. Shih, Argininosuccinate lyase deficiency: longterm
CE
[11]
PT
45.
outcome of 13 patients detected by newborn screening Molecular genetics and metabolism 98
[12]
AC
(2009) 273-277.
M. Marble, R.R. McGoey, E. Mannick, B. Keats, S.S. Ng, S. Deputy, H. Gereighty, E.
Schmidt-Sommerfeld, Living related liver transplant in a patient with argininosuccinic aciduria and cirrhosis: metabolic follow-up Journal of pediatric gastroenterology and nutrition 46 (2008) 453-456. [13]
J.V. Leonard, P.J. McKiernan, The role of liver transplantation in urea cycle disorders
21
ACCEPTED MANUSCRIPT Molecular genetics and metabolism 81 Suppl 1 (2004) S74-78. [14]
J.M. Saudubray, G. Touati, P. Delonlay, P. Jouvet, C. Narcy, J. Laurent, D. Rabier, P.
Kamoun, D. Jan, Y. Revillon, Liver transplantation in urea cycle disorders European journal of pediatrics 158 Suppl 2 (1999) S55-59. E. Robberecht, S. Maesen, A. Jonckheere, S. Van Biervliet, D. Carton, Successful liver
T
[15]
IP
transplantation for argininosuccinate lyase deficiency (ASLD) Journal of inherited metabolic
[16]
CR
disease 29 (2006) 184-185.
T. Newnham, W. Hardikar, K. Allen, R.M. Wellard, C. Hamilton, P. Angus, R. Jones, A.
US
Boneh, Liver transplantation for argininosuccinic aciduria: clinical, biochemical, and metabolic
AN
outcome Liver transplantation : official publication of the American Association for the Study of Liver Diseases and the International Liver Transplantation Society 14 (2008) 41-45. L. Wang, H. Morizono, J. Lin, P. Bell, D. Jones, D. McMenamin, H. Yu, M.L. Batshaw,
M
[17]
ED
J.M. Wilson, Preclinical evaluation of a clinical candidate AAV8 vector for ornithine transcarbamylase (OTC) deficiency reveals functional enzyme from each persisting vector
R.J. Chandler, T.N. Tarasenko, K. Cusmano-Ozog, Q. Sun, V.R. Sutton, C.P. Venditti,
CE
[18]
PT
genome Molecular genetics and metabolism 105 (2012) 203-211.
P.J. McGuire, Liver-directed adeno-associated virus serotype 8 gene transfer rescues a lethal
[19]
AC
murine model of citrullinemia type 1 Gene therapy 20 (2013) 1188-1191. L. Wang, H. Wang, H. Morizono, P. Bell, D. Jones, J. Lin, D. McMenamin, H. Yu, M.L.
Batshaw, J.M. Wilson, Sustained correction of OTC deficiency in spf( ash) mice using optimized self-complementary AAV2/8 vectors Gene Ther 19 (2012) 404-410. [20]
C.Y. Kok, S.C. Cunningham, K.H. Carpenter, A.P. Dane, S.M. Siew, G.J. Logan, P.W.
Kuchel, I.E. Alexander, Adeno-associated virus-mediated rescue of neonatal lethality in
22
ACCEPTED MANUSCRIPT argininosuccinate synthetase-deficient mice Mol. Ther. 21 (2013) 1823-1831. [21]
E.K. Lee, C. Hu, R. Bhargava, R. Ponnusamy, H. Park, S. Novicoff, N. Rozengurt, B.
Marescau, P. De Deyn, D. Stout, L. Schlichting, W.W. Grody, S.D. Cederbaum, G.S. Lipshutz, AAV-based gene therapy prevents neuropathology and results in normal cognitive development
Lipshutz, Minimal
ureagenesis
is
IP
C. Hu, D.S. Tai, H. Park, G. Cantero, E. Chan, M. Yudkoff, S.D. Cederbaum, G.S. necessary for survival in the murine model of
CR
[22]
T
in the hyperargininemic mouse Gene Ther. 20 (2013) 785-796.
hyperargininemia treated by AAV-based gene therapy Gene Ther. 22 (2015) 111-115. L. Wang, P. Bell, H. Morizono, Z. He, E. Pumbo, H. Yu, J. White, M.L. Batshaw, J.M.
US
[23]
AN
Wilson, AAV gene therapy corrects OTC deficiency and prevents liver fibrosis in aged OTCknock out heterozygous mice Mol. Genet. Metab. 120 (2017) 299-305. I.K. Kim, A.K. Niemi, C. Krueger, C.A. Bonham, W. Concepcion, T.M. Cowan, G.M.
M
[24]
ED
Enns, C.O. Esquivel, Liver transplantation for urea cycle disorders in pediatric patients: a singlecenter experience Pediatr. Transplant. 17 (2013) 158-167. M. Lock, M. Alvira, L.H. Vandenberghe, A. Samanta, J. Toelen, Z. Debyser, J.M.
PT
[25]
CE
Wilson, Rapid, simple, and versatile manufacturing of recombinant adeno-associated viral vectors at scale Hum Gene Ther 21 (2010) 1259-1271. P. Bell, A.D. Moscioni, R.J. McCarter, D. Wu, G. Gao, A. Hoang, J.C. Sanmiguel, X.
AC
[26]
Sun, N.A. Wivel, S.E. Raper, E.E. Furth, M.L. Batshaw, J.M. Wilson, Analysis of Tumors Arising in Male B6C3F1 Mice with and without AAV Vector Delivery to Liver Molecular therapy : the journal of the American Society of Gene Therapy 14 (2006) 34-44. [27]
V. Reid Sutton, Y. Pan, E.C. Davis, W.J. Craigen, A mouse model of argininosuccinic
aciduria: biochemical characterization Molecular genetics and metabolism 78 (2003) 11-16.
23
ACCEPTED MANUSCRIPT [28]
S.C. Cunningham, A. Spinoulas, K.H. Carpenter, B. Wilcken, P.W. Kuchel, I.E.
Alexander, AAV2/8-mediated correction of OTC deficiency is robust in adult but not neonatal Spf(ash) mice Molecular therapy : the journal of the American Society of Gene Therapy 17 (2009) 1340-1346. L. Wang, H. Wang, P. Bell, D. McMenamin, J.M. Wilson, Hepatic gene transfer in
T
[29]
J.A. Greig, J.M.L. Nordin, C. Draper, P. Bell, J.M. Wilson, AAV8 Gene Therapy
CR
[30]
IP
neonatal mice by adeno-associated virus serotype 8 vector Hum Gene Ther 23 (2012) 533-539.
Rescues the Newborn Phenotype of a Mouse Model of Crigler-Najjar Hum Gene Ther (2018). A.M. Davidoff, C.Y. Ng, J. Zhou, Y. Spence, A.C. Nathwani, Sex significantly
US
[31]
AN
influences transduction of murine liver by recombinant adeno-associated viral vectors through an androgen-dependent pathway Blood 102 (2003) 480-488. M. Corti, M. Elder, D. Falk, L. Lawson, B. Smith, S. Nayak, T. Conlon, N. Clement, K.
M
[32]
ED
Erger, E. Lavassani, M. Green, P. Doerfler, R. Herzog, B. Byrne, B-Cell Depletion is Protective Against Anti-AAV Capsid Immune Response: A Human Subject Case Study Molecular therapy.
F. Mingozzi, Y. Chen, S.L. Murphy, S.C. Edmonson, A. Tai, S.D. Price, M.E. Metzger,
CE
[33]
PT
Methods & clinical development 1 (2014).
S. Zhou, J.F. Wright, R.E. Donahue, C.E. Dunbar, K.A. High, Pharmacological modulation of
AC
humoral immunity in a nonhuman primate model of AAV gene transfer for hemophilia B Molecular therapy : the journal of the American Society of Gene Therapy 20 (2012) 1410-1416. [34]
Q.R. Rogers, R.A. Freedland, R.A. Symmons, In vivo synthesis and utilization of
arginine in the rat Am J Physiol 223 (1972) 236-240. [35]
A. Nagasaki, T. Gotoh, M. Takeya, Y. Yu, M. Takiguchi, H. Matsuzaki, K. Takatsuki, M.
Mori, Coinduction of nitric oxide synthase, argininosuccinate synthetase, and argininosuccinate
24
ACCEPTED MANUSCRIPT lyase in lipopolysaccharide-treated rats. RNA blot, immunoblot, and immunohistochemical analyses J Biol Chem 271 (1996) 2658-2662. [36]
S.C. Nagamani, P.M. Campeau, O.A. Shchelochkov, M.H. Premkumar, K. Guse, N.
Brunetti-Pierri, Y. Chen, Q. Sun, Y. Tang, D. Palmer, A.K. Reddy, L. Li, T.C. Slesnick, D.I.
T
Feig, S. Caudle, D. Harrison, L. Salviati, J.C. Marini, N.S. Bryan, A. Erez, B. Lee, Nitric-oxide
IP
supplementation for treatment of long-term complications in argininosuccinic aciduria Am J
[37]
CR
Hum Genet 90 (2012) 836-846.
R. Wiest, R.J. Groszmann, The paradox of nitric oxide in cirrhosis and portal
AC
CE
PT
ED
M
AN
US
hypertension: too much, not enough Hepatology 35 (2002) 478-491.
25
ACCEPTED MANUSCRIPT Figure Legends
Figure 1. AAV8 gene therapy extends survival of an ASA hypomorphic mouse model. (A) A survival curve of ASA hypomorphic mice is shown (n = 81). (B) ASA hypomorphic mice
T
were injected IV via the temporal vein with 10 10 GC/mouse (n=5) or 1011 GC/mouse (n=6) of
IP
AAV8.TBG.hASLco and monitored for survival. Comparisons to uninjected ASA hypomorphic
CR
mice (solid line) are shown. The weight of female (C) and male (D) ASA hypomorphic mice administered with 1011 GC/mouse of AAV8.TBG.hASLco was monitored, and compared to
US
wild-type (WT) mice and ASA hypomorphic mice that did not receive gene therapy. Error bars
AN
= standard error of the mean (SEM); **p < 0.01, ***p <0.001.
M
Figure 2. ASL protein expression in the liver of newborn injected mice.
ED
ASA hypomorphic mice were injected IV via the temporal vein with 10 10 GC/mouse (n=5) or 1011 GC/mouse (n=6) of AAV8.TBG.hASLco.
Mice were necropsied after cohort-specific
PT
median survival was reached. Livers were harvested and IHC was performed for detection of the
CE
ASL protein, and compared to wild-type (WT), heterozygous (Het), and untreated ASA hypomorphic mice.
Central veins are denoted by asterisks.
Representative sections from
AC
different mice in each cohort are shown.
Figure 3.
Sex difference in survival and efficacy of AAV8 gene therapy in ASA
hypomorphic mice. ASA hypomorphic mice were injected IV via the retro orbital vein on P30 with 1013 or 6x1013 GC/kg of AAV8.TBG.hASLco (n=8/group). Mice were monitored for survival (A) and weight
26
ACCEPTED MANUSCRIPT (B). Comparisons to uninjected ASA hypomorphic mice are shown (female, n=3; male, n=4). Mice were bled and plasma was analyzed for the liver transaminases AST (C) and ALT (D). Comparison of values to uninjected ASA hypomorphic mice (no vector) at the same time point
T
are shown (n=5). Error bars = SEM; **p < 0.01, ***p <0.001.
IP
Figure 4. High-dose AAV8 gene therapy corrects liver metabolism in ASA hypomorphic
CR
mice.
ASA hypomorphic mice were injected IV via the orbital vein on P30 with 1013 or 6x1013 GC/kg
US
of AAV8.TBG.hASLco (n=8/group). Mice were bled and plasma was analyzed for components
AN
of the urea cycle, including (A) citrulline, (B) ASA, and (C) arginine. Comparisons to levels in wild-type (WT) mice are shown (n=22), ASA hypomorph control (n=12). Error bars = SEM; ns,
ED
M
not significant; *p < 0.05, **p < 0.01, ***p <0.001.
Figure 5. Detection of ASL protein expression and genome copies in the liver of adolescent
PT
injected mice.
CE
ASA hypomorphic mice were injected IV via the retro orbital vein at P30 with 1013 (n=8/sex) or 6x1013 GC/kg of AAV8.TBG.hASLco (n=8/group). (A) Livers were harvested and IHC was
AC
performed for detection of the ASL protein. Each section is a representative sample from a different mouse within each cohort.
Central veins are denoted by asterisks.
(B) Liver
homogenates were assayed for ASL activity compared to wild-type levels (data presented as percentage of wild-type activity). Analysis of activity was performed by a one-way ANOVA with Dunnett’s multiple comparison test comparing each group to uninjected ASA hypomorphic mice (no vector). (C) Following extraction of DNA from liver, vector GCs were determined.
27
ACCEPTED MANUSCRIPT Error bars = SEM; ***p < 0.001.
Figure 6.
Histopathologic findings in the liver of a mouse model of ASL administered
either a low (1x1013 GC) or high dose (6x1013 GC) of AAV.
T
ASA hypomorphic mice were injected IV via the retro orbital vein at P30 with 10 13 (n=8/sex) or
IP
6x1013 GC/kg of AAV8.TBG.hASLco (n=8/group). Livers were harvested and H&E and Sirius
CR
red staining were performed for observation of histopathology. Findings from each category were ranked from 0-5 for hepatocellular changes with 0 being no finding, 5 being marked
US
karyocytomegaly (>90% of hepatocytes within a lobule) with lobular collapse and numerous
AN
single-cell necrosis, and 1-3 denoting increasing severity.
M
Figure 7. Histopathologic findings in the liver of a mouse model of ASL administered with
ED
either a low (1x1013 GC) or high dose (6x1013 GC) of AAV. The images depict representative trends of the histopathological findings for each dose group.
PT
(A) A male mouse received a low dose of vector and exhibited grade 4 hepatocellular changes
CE
characterized by severe karyocytomegaly (51-90% of hepatocytes within a lobule) with extensive hepatocellular dissociation and frequent individual cell necrosis; however, generally
AC
normal hepatocellular architecture was maintained. This animal also had oval cell (grade 3) and bile duct (grade 1) hyperplasia.
(B) A female mouse also received a low dose of vector;
however, hepatocellular changes were less severe (grade 2) characterized by mild karyocytomegaly (< 10% of hepatocytes within a lobule) with normal hepatic architecture and no individual cell necrosis.
This animal had no oval or bile duct hyperplasia.
(C) The
histopathologic findings were similar in a female mouse administered with a high dose of vector.
28
ACCEPTED MANUSCRIPT (D) However, other animals in this dose group had no significant liver findings, as depicted in the displayed mouse. H&E, Scale bar = 100 µm.
Figure 8.
Variable fibrosis was observed in the liver of a mouse model of ASL
T
administered with either a low (1x1013 GC) or high dose (6x1013 GC) of AAV.
IP
Sirius red- and H&E-stained liver sections were scored based on location and amount of hepatic
CR
infiltration. The severe cases (grade 3) were mostly in the low-dose group and were clearly visible on H&E staining. (A) In a male mouse from the low-dose cohort, the liver had regions of
US
bridging fibrosis between portal and occasionally centrilobular areas with architectural disruption
AN
and individualization of hepatocytes. The presence of hepatocellular mitotic figures, indicative of regeneration, was frequently observed in these areas (inset, scale bar = 20 pixel). In these
M
cases, the fibrosis was frequently accompanied by oval cell and bile duct hyperplasia. Sirius red
ED
staining highlighted the fibrosis in less severely affected sections. (B) A male mouse in the lowdose group exhibited regional bridging portal fibrosis (grade 2) with no architectural distortion,
PT
(C) whereas only periportal fibrosis (grade 1) was observed in a female mouse from the high-
CE
dose group. (D) No fibrosis was observed in another female mouse from the high-dose group.
AC
Sirius red, Scale bar = 100 µm.
29
Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7
Figure 8