Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Scott D. Bercury is active.

Publication


Featured researches published by Scott D. Bercury.


Molecular Therapy | 2009

Glycoengineered Acid α-Glucosidase With Improved Efficacy at Correcting the Metabolic Aberrations and Motor Function Deficits in a Mouse Model of Pompe Disease

Yunxiang Zhu; Ji-Lei Jiang; Nathan K. Gumlaw; Jinhua Zhang; Scott D. Bercury; Robin J. Ziegler; Karen Lee; Mariko Kudo; William M. Canfield; Timothy Edmunds; Canwen Jiang; Robert J. Mattaliano; Seng H. Cheng

Improving the delivery of therapeutics to disease-affected tissues can increase their efficacy and safety. Here, we show that chemical conjugation of a synthetic oligosaccharide harboring mannose 6-phosphate (M6P) residues onto recombinant human acid alpha-glucosidase (rhGAA) via oxime chemistry significantly improved its affinity for the cation-independent mannose 6-phosphate receptor (CI-MPR) and subsequent uptake by muscle cells. Administration of the carbohydrate-remodeled enzyme (oxime-neo-rhGAA) into Pompe mice resulted in an approximately fivefold higher clearance of lysosomal glycogen in muscles when compared to the unmodified counterpart. Importantly, treatment of immunotolerized Pompe mice with oxime-neo-rhGAA translated to greater improvements in muscle function and strength. Treating older, symptomatic Pompe mice also reduced tissue glycogen levels but provided only modest improvements in motor function. Examination of the muscle pathology suggested that the poor response in the older animals might have been due to a reduced regenerative capacity of the skeletal muscles. These findings lend support to early therapeutic intervention with a targeted enzyme as important considerations in the management of Pompe disease.Improving the delivery of therapeutics to disease-affected tissues can increase their efficacy and safety. Here, we show that chemical conjugation of a synthetic oligosaccharide harboring mannose 6-phosphate (M6P) residues onto recombinant human acid α-glucosidase (rhGAA) via oxime chemistry significantly improved its affinity for the cation-independent mannose 6-phosphate receptor (CI-MPR) and subsequent uptake by muscle cells. Administration of the carbohydrate-remodeled enzyme (oxime-neo-rhGAA) into Pompe mice resulted in an approximately fivefold higher clearance of lysosomal glycogen in muscles when compared to the unmodified counterpart. Importantly, treatment of immunotolerized Pompe mice with oxime-neo-rhGAA translated to greater improvements in muscle function and strength. Treating older, symptomatic Pompe mice also reduced tissue glycogen levels but provided only modest improvements in motor function. Examination of the muscle pathology suggested that the poor response in the older animals might have been due to a reduced regenerative capacity of the skeletal muscles. These findings lend support to early therapeutic intervention with a targeted enzyme as important considerations in the management of Pompe disease.


Molecular Therapy | 2010

Preexisting Immunity and Low Expression in Primates Highlight Translational Challenges for Liver-directed AAV8-mediated Gene Therapy

Gregory D Hurlbut; Robin J. Ziegler; Jennifer B. Nietupski; Joseph Foley; Lisa Woodworth; Elizabeth Meyers; Scott D. Bercury; Nilesh Pande; David W. Souza; Mark Bree; Michael Lukason; John Marshall; Seng H. Cheng; Ronald K. Scheule

Liver-directed gene therapy with adeno-associated virus (AAV) vectors effectively treats mouse models of lysosomal storage diseases (LSDs). We asked whether these results were likely to translate to patients. To understand to what extent preexisting anti-AAV8 antibodies could impede AAV8-mediated liver transduction in primates, commonly preexposed to AAV, we quantified the effects of preexisting antibodies on liver transduction and subsequent transgene expression in mouse and nonhuman primate (NHP) models. Using the highest viral dose previously reported in a clinical trial, passive transfer of NHP sera containing relatively low anti-AAV8 titers into mice blocked liver transduction, which could be partially overcome by increasing vector dose tenfold. Based on this and a survey of anti-AAV8 titers in 112 humans, we predict that high-dose systemic gene therapy would successfully transduce liver in >50% of human patients. However, although high-dose AAV8 administration to mice and monkeys with equivalent anti-AAV8 titers led to comparable liver vector copy numbers, the resulting transgene expression in primates was ~1.5-logs lower than mice. This suggests vector fate differs in these species and that strategies focused solely on overcoming preexisting vector-specific antibodies may be insufficient to achieve clinically meaningful expression levels of LSD genes using a liver-directed gene therapy approach in patients.


Molecular Therapy | 2007

Correction of the Biochemical and Functional Deficits in Fabry Mice Following AAV8–mediated Hepatic Expression of α-galactosidase A

Robin J. Ziegler; Maribeth Cherry; Christine M. Barbon; Chester Li; Scott D. Bercury; Donna Armentano; Robert J. Desnick; Seng H. Cheng

The advent of novel adeno-associated virus (AAV) serotype vectors with higher transduction activity has encouraged a re-evaluation of the merits of this delivery platform for a variety of diseases. We report here that administration of a recombinant AAV8-based serotype vector encoding human α-galactosidase A into Fabry mice facilitated more rapid and significantly higher levels of production of the enzyme than an AAV2 vector. This translated into improved clearance of globotriaosylceramide, the glycosphingolipid that accumulates in the lysosomes of affected Fabry cells, and to correction of the peripheral neuropathy shown associated with this disease. The higher levels of α-galactosidase A expression also allowed for a more rapid induction of immunotolerance to the enzyme. Recombinant AAV8 vectors that facilitated hepatic-restricted expression of high levels of α-galactosidase A conferred immunotolerance to the expressed enzyme as early as 30 days post-treatment. Animals expressing lower levels of the hydrolase, such as those treated with an AAV2-based vector or with lower doses of the AAV8-based vector, were also able to develop immunotolerance, but only after a more extended time period. Adoptive transfer of T cells isolated from the spleens of immunotolerized mice suppressed the formation of antibodies in naïve recipient animals, suggesting the possible role of regulatory T cells in effecting this state.


Human Gene Therapy | 2008

Ability of Adeno-Associated Virus Serotype 8-Mediated Hepatic Expression of Acid α-Glucosidase to Correct the Biochemical and Motor Function Deficits of Presymptomatic and Symptomatic Pompe Mice

Robin J. Ziegler; Scott D. Bercury; Jonathan A. Fidler; Michael A. Zhao; Joseph Foley; Tatyana V. Taksir; Susan Ryan; Bradley L. Hodges; Ronald K. Scheule; Lamya S. Shihabuddin; Seng H. Cheng

The availability of a murine model of Pompe disease has enabled an evaluation of the relative merits of various therapeutic paradigms, including gene therapy. We report here that administration of a recombinant adeno-associated virus serotype 8 (AAV8) vector (AAV8/DC190-GAA) encoding human acid alpha-glucosidase (GAA) into presymptomatic Pompe mice resulted in nearly complete correction of the lysosomal storage of glycogen in all the affected muscles. A relatively high dose of AAV8/DC190-GAA was necessary to attain a threshold level of GAA for inducing immunotolerance to the expressed enzyme and for correction of muscle function, coordination, and strength. Administration of AAV8/DC190-GAA into older Pompe mice with overt disease manifestations was also effective at correcting the lysosomal storage abnormality. However, these older mice exhibited only marginal improvements in motor function and no improvement in muscle strength. Examination of histologic sections showed evidence of skeletal muscle degeneration and fibrosis in aged Pompe mice whose symptoms were abated or rescued by early but not late treatment with AAV8/DC190-GAA. These results suggest that AAV8-mediated hepatic expression of GAA was effective at addressing the biochemical and functional deficits in Pompe mice. However, early therapeutic intervention is required to maintain significant muscle function and should be an important consideration in the management and treatment of Pompe disease.


PLOS ONE | 2011

Iminosugar-based inhibitors of glucosylceramide synthase increase brain glycosphingolipids and survival in a mouse model of Sandhoff disease

Karen M. Ashe; Dinesh S. Bangari; Lingyun Li; Mario A. Cabrera-Salazar; Scott D. Bercury; Jennifer B. Nietupski; Christopher G.F. Cooper; Johannes M. F. G. Aerts; Edward R. Lee; Diane P. Copeland; Seng H. Cheng; Ronald K. Scheule; John Marshall

The neuropathic glycosphingolipidoses are a subgroup of lysosomal storage disorders for which there are no effective therapies. A potential approach is substrate reduction therapy using inhibitors of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide and related glycosphingolipids that accumulate in the lysosomes. Genz-529468, a blood-brain barrier-permeant iminosugar-based GCS inhibitor, was used to evaluate this concept in a mouse model of Sandhoff disease, which accumulates the glycosphingolipid GM2 in the visceral organs and CNS. As expected, oral administration of the drug inhibited hepatic GM2 accumulation. Paradoxically, in the brain, treatment resulted in a slight increase in GM2 levels and a 20-fold increase in glucosylceramide levels. The increase in brain glucosylceramide levels might be due to concurrent inhibition of the non-lysosomal glucosylceramidase, Gba2. Similar results were observed with NB-DNJ, another iminosugar-based GCS inhibitor. Despite these unanticipated increases in glycosphingolipids in the CNS, treatment nevertheless delayed the loss of motor function and coordination and extended the lifespan of the Sandhoff mice. These results suggest that the CNS benefits observed in the Sandhoff mice might not necessarily be due to substrate reduction therapy but rather to off-target effects.


PLOS ONE | 2012

Systemic delivery of a glucosylceramide synthase inhibitor reduces CNS substrates and increases lifespan in a mouse model of type 2 Gaucher disease.

Mario A. Cabrera-Salazar; Matthew DeRiso; Scott D. Bercury; Lingyun Li; John Lydon; William Weber; Nilesh Pande; Mandy Cromwell; Diane P. Copeland; John P. Leonard; Seng H. Cheng; Ronald K. Scheule

Neuropathic Gaucher disease (nGD), also known as type 2 or type 3 Gaucher disease, is caused by a deficiency of the enzyme glucocerebrosidase (GC). This deficiency impairs the degradation of glucosylceramide (GluCer) and glucosylsphingosine (GluSph), leading to their accumulation in the brains of patients and mouse models of the disease. These accumulated substrates have been thought to cause the severe neuropathology and early death observed in patients with nGD and mouse models. Substrate accumulation is evident at birth in both nGD mouse models and humans affected with the most severe type of the disease. Current treatment of non-nGD relies on the intravenous delivery of recombinant human glucocerebrosidase to replace the missing enzyme or the administration of glucosylceramide synthase inhibitors to attenuate GluCer production. However, the currently approved drugs that use these mechanisms do not cross the blood brain barrier, and thus are not expected to provide a benefit for the neurological complications in nGD patients. Here we report the successful reduction of substrate accumulation and CNS pathology together with a significant increase in lifespan after systemic administration of a novel glucosylceramide synthase inhibitor to a mouse model of nGD. To our knowledge this is the first compound shown to cross the blood brain barrier and reduce substrates in this animal model while significantly enhancing its lifespan. These results reinforce the concept that systemically administered glucosylceramide synthase inhibitors could hold enhanced therapeutic promise for patients afflicted with neuropathic lysosomal storage diseases.


Experimental Neurology | 2010

Intracerebroventricular delivery of glucocerebrosidase reduces substrates and increases lifespan in a mouse model of neuronopathic Gaucher disease

Mario A. Cabrera-Salazar; Scott D. Bercury; Robin J. Ziegler; John Marshall; Bradley L. Hodges; Wei-Lien Chuang; Joshua Pacheco; Lingyun Li; Seng H. Cheng; Ronald K. Scheule

Gaucher disease is caused by a deficit in the enzyme glucocerebrosidase. As a consequence, degradation of the glycolipids glucosylceramide (GluCer) and glucosylsphingosine (GluSph) is impaired, and their subsequent buildup can lead to significant pathology and early death. Type 1 Gaucher patients can be treated successfully with intravenous replacement enzyme, but this enzyme does not reach the CNS and thus does not ameliorate the neurological involvement in types 2 and 3 Gaucher disease. As one potential approach to treating these latter patients, we have evaluated intracerebroventricular (ICV) administration of recombinant human glucocerebrosidase (rhGC) in a mouse model of neuronopathic Gaucher disease. ICV administration resulted in enzyme distribution throughout the brain and alleviated neuropathology in multiple brain regions of this mouse model. Treatment also resulted in dose-dependent decreases in GluCer and GluSph and significantly extended survival. To evaluate the potential of continuous enzyme delivery, a group of animals was treated ICV with an adeno-associated viral vector encoding hGC and resulted in a further extension of survival. These data suggest that ICV administration of rhGC may represent a potential therapeutic approach for type 2/3 Gaucher patients. Preclinical evaluation in larger animals will be needed to ascertain the translatability of this approach to the clinic.


Experimental Neurology | 2011

Distribution of acid sphingomyelinase in rodent and non-human primate brain after intracerebroventricular infusion.

Robin J. Ziegler; Ernesto A. Salegio; James Dodge; John Bringas; Christopher M. Treleaven; Scott D. Bercury; Thomas J. Tamsett; Lamya S. Shihabuddin; Piotr Hadaczek; Massimo S. Fiandaca; Krystof S. Bankiewicz; Ronald K. Scheule

One treatment approach for lysosomal storage diseases (LSDs) is the systemic infusion of recombinant enzyme. Although this enzyme replacement is therapeutic for the viscera, many LSDs have central nervous system (CNS) components that are not adequately treated by systemic enzyme infusion. Direct intracerebroventricular (ICV) infusion of a high concentration of recombinant human acid sphingomyelinase (rhASM) into the CNS over a prolonged time frame (hours) has shown therapeutic efficacy in a mouse model of Niemann-Pick A (NP/A) disease. To evaluate whether such an approach would translate to a larger brain, rhASM was infused into the lateral ventricles of both rats and Rhesus macaques, and the resulting distribution of enzyme characterized qualitatively and quantitatively. In both species, ICV infusion of rhASM resulted in parenchymal distribution of enzyme at levels that were therapeutic in the NP/A mouse model. Enzyme distribution was global in nature and exhibited a relatively steep gradient from the cerebrospinal fluid compartment to the inner parenchyma. Additional optimization of an ICV delivery approach may provide a therapeutic option for LSDs with neurologic involvement.


Molecular Therapy | 2010

Evaluation of Systemic Follistatin as an Adjuvant to Stimulate Muscle Repair and Improve Motor Function in Pompe Mice

Joseph Foley; Scott D. Bercury; Patrick Finn; Seng H. Cheng; Ronald K. Scheule; Robin J. Ziegler

Due to the lack of acid alpha-glucosidase (GAA) activity, Pompe mice develop glycogen storage pathology and progressive skeletal muscle dysfunction with age. Applying either gene or enzyme therapy to reconstitute GAA levels in older, symptomatic Pompe mice effectively reduces glycogen storage in skeletal muscle but provides only modest improvements in motor function. As strategies to stimulate muscle hypertrophy, such as by myostatin inhibition, have been shown to improve muscle pathology and strength in mouse models of muscular dystrophy, we sought to determine whether these benefits might be similarly realized in Pompe mice. Administration of a recombinant adeno-associated virus serotype 8 vector encoding follistatin, an inhibitor of myostatin, increased muscle mass and strength but only in Pompe mice that were treated before 10 months of age. Younger Pompe mice showed significant muscle fiber hypertrophy in response to treatment with follistatin, but maximal gains in muscle strength were achieved only when concomitant GAA administration reduced glycogen storage in the affected muscles. Despite increased grip strength, follistatin treatment failed to improve rotarod performance. These findings highlight the importance of treating Pompe skeletal muscle before pathology becomes irreversible, and suggest that adjunctive therapies may not be effective without first clearing skeletal muscle glycogen storage with GAA.


Molecular Therapy | 2005

245. AAV2/8-Mediated Efficacy and Immune Tolerance in a Pompe Disease Mouse Model

Scott D. Bercury; Robin J. Ziegler; Christine M. Barbon; David W. Souza; Donna Armentano; Beth L. Thurberg; Tatyana V. Taksir; H. Do; M. Trout; N. Raben; Seng H. Cheng

Pompe disease is an inherited metabolic disorder caused by a deficiency of the lysosomal enzyme acid -glucosidase (GAA). The disease is characterized by accumulation of glycogen in various tissues, including the central and peripheral nervous systems, but the pathology most dramatically affects skeletal and cardiac muscles. Severely affected patients present within the first year of life with hypotonia and cardiomyopathy, resulting in death from cardiopulmonary failure. To assess the utility of gene therapy for Pompe disease, we generated recombinant AAV8-based serotype vectors encoding for GAA under the transcriptional control of a liver-restricted promoter (DC190). Systemic administration of 5X1011 drp of AAV8/DC190-GAA into Pompe mice generated high levels (25-30 g/ml) of GAA in the serum that persisted for 6 months post-administration. GAA activity was also detected in several muscle groups isolated from the AAV8-treated mice, indicating that GAA was efficiently taken up into these tissues from circulation. Glycogen storage was completely corrected in the heart and diaphragm and reduced by 50% in the skeletal muscles as early as 4 weeks post-treatment. All tissues were completely cleared of glycogen storage by 10 weeks, and remained at baseline at the 6-month time point. Histomorphometric analysis of PAS-stained tissues confirmed that glycogen deposits were effectively cleared from the affected tissues. Histopathology observed in the dorsal root ganglia of Pompe mice was also normalized suggesting that treatment may impact the peripheral nerve disease. Consistent with previous studies using the DC190 promoter, no antibodies to the human GAA were detected in the sera of AAV8/DC190-GAA-treated mice. Furthermore, no antibodies were generated following two subsequent challenges with recombinant GAA emulsified in Complete Freunds adjuvant, suggesting the AAV8/DC190-GAA-treated mice were immune tolerized. Immune responses were also assessed at the cellular level by assaying lymphocyte proliferation and IFN secretion using cells harvested from the spleens of the treated mice. Cells from the AAV8/DC190-GAA-treated mice showed an attenuated response to antigenic stimulation compared to controls. These data support the continued evaluation of AAV8-mediated gene therapy as an approach to treat the systemic manifestations of Pompe disease.

Collaboration


Dive into the Scott D. Bercury's collaboration.

Researchain Logo
Decentralizing Knowledge