Network


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

Hotspot


Dive into the research topics where Qiuming Chu is active.

Publication


Featured researches published by Qiuming Chu.


Human Gene Therapy | 2002

Development of Catheter-Based Procedures for Transducing the Isolated Rabbit Liver with Plasmid DNA

Simon J. Eastman; Kevin M. Baskin; Bradley L. Hodges; Qiuming Chu; Amy L. Gates; Rebecca Dreusicke; Scott C. Anderson; Ronald K. Scheule

Rapid systemic injection of naked plasmid DNA (pDNA) in a large volume into a mouse tail vein has been shown to result in a high level of gene expression in the liver. However, the potential therapeutic benefit to humans embodied in hydrodynamic transfection of the liver cannot be realized until a clinically viable method for gene delivery is developed. In light of this fact, we have devised and evaluated several methods for delivering pDNA to the isolated rabbit liver using minimally invasive catheter-based techniques. Using a lobar technique, pDNA was delivered hydrodynamically to an isolated hepatic lobe using a balloon occlusion balloon catheter to occlude a selected hepatic vein. A whole organ technique was used wherein the entire hepatic venous system was isolated and the pDNA solution injected hydrodynamically into the vena cava between two balloons used to block hepatic venous outflow. Lobar delivery of a plasmid encoding a secreted alkaline phosphatase (SEAP) reporter gene resulted in significant levels of transgene product in the serum. A nonsecreted transgene product, chloramphenicol acetyltransferase (CAT), showed the highest levels of expression in the injected lobe distal to the injection site. Compared to lobar delivery, whole organ delivery yielded much higher serum levels of SEAP expression and a significantly broader hepatic parenchymal distribution of CAT expression. These preliminary studies suggest that catheter-mediated hydrodynamic delivery of pDNA to the isolated liver may provide a method for human gene therapy that is both therapeutically significant and clinically practicable.


Molecular Genetics and Metabolism | 2010

Inhibition of glycogen biosynthesis via mTORC1 suppression as an adjunct therapy for Pompe disease.

Karen M. Ashe; Kristin M. Taylor; Qiuming Chu; Elizabeth Meyers; Allen Ellis; Varvara Jingozyan; Katherine W. Klinger; Patrick Finn; Christopher G.F. Cooper; Wei-Lien Chuang; John Marshall; John M. McPherson; Robert J. Mattaliano; Seng H. Cheng; Ronald K. Scheule; Rodney J. Moreland

Pompe disease, also known as glycogen storage disease (GSD) type II, is caused by deficiency of lysosomal acid alpha-glucosidase (GAA). The resulting glycogen accumulation causes a spectrum of disease severity ranging from a rapidly progressive course that is typically fatal by 1-2years of age to a more slowly progressive course that causes significant morbidity and early mortality in children and adults. Recombinant human GAA (rhGAA) improves clinical outcomes with variable results. Adjunct therapy that increases the effectiveness of rhGAA may benefit some Pompe patients. Co-administration of the mTORC1 inhibitor rapamycin with rhGAA in a GAA knockout mouse reduced muscle glycogen content more than rhGAA or rapamycin alone. These results suggest mTORC1 inhibition may benefit GSDs that involve glycogen accumulation in muscle.


Molecular Therapy | 2008

Systemic Insulin-like Growth Factor-1 Reverses Hypoalgesia and Improves Mobility in a Mouse Model of Diabetic Peripheral Neuropathy

Qiuming Chu; Rod Moreland; Nelson S. Yew; Joseph Foley; Robin J. Ziegler; Ronald K. Scheule

Peripheral neuropathy is a particularly debilitating complication of both type 1 and type 2 diabetes characterized by sensory and motor neuron damage and decreased circulating levels of insulin-like growth factor 1 (IGF-1). Quite often, an early hyperalgesia is followed by hypoalgesia and muscle weakness. Hypoalgesia can lead to significant morbidity for which there is no current treatment. Hyperglycemic, streptozotocin (STZ)-induced rodent models reproduce these symptoms. We investigated whether increasing systemic IGF-1 could improve neuronal function in hyper- and hypoalgesic STZ-treated mice. Increased circulating levels of IGF-1 were achieved by delivering a plasmid or adeno-associated viral (AAV) vector bearing mouse IGF-1 to the liver. Treating mice in the hyperalgesia stage prevented later hypoalgesia. Treating mice in the hypoalgesia stage reversed existing hypoalgesia. This latter effect could be seen by merely restoring IGF-1 serum levels to normalcy, which was possible to achieve by IGF-1 gene therapy or insulin treatment. Sensory nerve functional correction was seen to be correlated with attenuated Schwann cell vacuolization and demyelination in peripheral sensory nerve fibers. A further increase in serum IGF-1 levels with gene therapy also improved motor function, consistent with the observed prevention of both muscle atrophy and peripheral motor nerve fiber demyelination. These results suggest that the restoration of systemic levels of IGF-1 may prove to be a highly effective therapeutic modality for treating diabetic peripheral neuropathy.


Molecular Therapy | 2011

Systemic administration of AAV8-α-galactosidase A induces humoral tolerance in nonhuman primates despite low hepatic expression.

Jennifer B. Nietupski; Gregory D Hurlbut; Robin J. Ziegler; Qiuming Chu; Bradley L. Hodges; Karen M. Ashe; Mark Bree; Seng H. Cheng; Richard J. Gregory; John Marshall; Ronald K. Scheule

In mice, liver-restricted expression of lysosomal enzymes from adeno-associated viral serotype 8 (AAV8) vectors results in reduced antibodies to the expressed proteins. To ask whether this result might translate to patients, nonhuman primates (NHPs) were injected systemically with AAV8 encoding α-galactosidase A (α-gal). As in mice, sustained expression in monkeys attenuated antibody responses to α-gal. However, this effect was not robust, and sustained α-gal levels were 1-2 logs lower than those achieved in male mice at the same vector dose. Because our mouse studies had shown that antibody levels were directly related to expression levels, several strategies were evaluated to increase expression in monkeys. Unlike mice, expression in monkeys did not respond to androgens. Local delivery to the liver, immune suppression, a self-complementary vector and pharmacologic approaches similarly failed to increase expression. While equivalent vector copies reached mouse and primate liver and there were no apparent differences in vector form, methylation or deamination, transgene expression was limited at the mRNA level in monkeys. These results suggest that compared to mice, transcription from an AAV8 vector in monkeys can be significantly reduced. They also suggest some current limits on achieving clinically useful antibody reduction and therapeutic benefit for lysosomal storage diseases using a systemic AAV8-based approach.


Molecular Therapy | 2010

Induction of immune tolerance to a therapeutic protein by intrathymic gene delivery.

Qiuming Chu; Rodney J. Moreland; Lan Gao; Kristin M. Taylor; Elizabeth Meyers; Seng H. Cheng; Ronald K. Scheule

The efficacy of recombinant enzyme therapy for genetic diseases is limited in some patients by the generation of a humoral immune response to the therapeutic protein. Inducing immune tolerance to the protein prior to treatment has the potential to increase therapeutic efficacy. Using an AAV8 vector encoding human acid α-glucosidase (hGAA), we have evaluated direct intrathymic injection for inducing tolerance. We have also compared the final tolerogenic states achieved by intrathymic and intravenous injection. Intrathymic vector delivery induced tolerance equivalent to that generated by intravenous delivery, but at a 25-fold lower dose, the thymic hGAA expression level was 10,000-fold lower than the liver expression necessary for systemic tolerance induction. Splenic regulatory T cells (Tregs) were apparent after delivery by both routes, but with different phenotypes. Intrathymic delivery resulted in Tregs with higher FoxP3, TGFβ, and IL-10 mRNA levels. These differences may account for the differences noted in splenic T cells, where only intravenous delivery appeared to inhibit their activation. Our results imply that different mechanisms may be operating to generate immune tolerance by intrathymic and intravenous delivery of an AAV vector, and suggest that the intrathymic route may hold promise for decreasing the humoral immune response to therapeutic proteins in genetic disease indications.


Molecular Therapy | 2005

531. Balloon Catheter-Mediated Hepatic Vein Delivery of a Viral Vector Mitigates Neutralization by Anti-Viral Antibodies and Results in Efficient Transduction of Rabbit Liver

Bradley L. Hodges; Kristin M. Taylor; Qiuming Chu; Samantha E. Scull; Rebecca G. Serriello; Scott C. Anderson; Ronald K. Scheule

The liver, and in particular, hepatocytes, represent an attractive target for both viral and non-viral gene transfer vectors. However, there are certain delivery issues associated with both of these vector systems that make their use in humans problematic. In particular, for viral vectors, the existence of anti-viral antibodies in the general population represents a potential barrier to their effective use in the clinic. Building upon our earlier success using balloon catheters to deliver plasmid DNA by way of the hepatic venous circulation to rabbit liver, we have used this same model to ask if we could use a similar approach to deliver a model viral vector. Specifically, we have compared a local, balloon catheter-mediated hepatic vein delivery protocol to systemic delivery of a CMV-driven adenoviral vector expressing b-galactosidase in terms of liver transduction efficiency, toxicity, and cell types transduced. We have also made this comparison in the presence of defined anti-AdV antibody titers in the recipient rabbits. In the naive animal, local balloon catheter-based delivery conferred an advantage in overall liver transduction when compared to systemic delivery of an identical dose of virus. In rabbits bearing anti-AdV antibody titers equivalent to those found in pooled human serum, this difference in expression between local and systemic delivery was even more striking. Importantly, in the presence of passively-administered anti-AdV antibodies, balloon-catheter mediated delivery resulted in expression levels that were comparable to those obtained by systemic delivery of an equivalent dose in a naive animal. Since in general, systemic delivery of AdV in naive animal models results in expression levels of secreted proteins regarded as therapeutic, the present results predict that retrograde delivery by a hepatic vein route using a balloon catheter should result in therapeutic levels of expression, even in the presence of average human levels of anti-AdV antibodies. Further support for this hypothesis is the finding that this local hepatic vein delivery approach resulted in the majority of expression originating from hepatocytes even in the passively immunized animals. In contrast, systemic delivery of AdV in passively immunized animals resulted in the majority of expression originating from non-hepatocytes. This latter finding implies that this local approach should help minimize the transduction of antigen-presenting cells, thereby reducing any immune consequences of viral transduction. Taken together, these data suggest that coupled with additional features such as a hepatocyte-specific expression cassette, balloon catheter viral-mediated transduction of the liver may represent a practical clinical approach for treating indications for which hepatocytes can be used as a depot for therapeutic protein production.


Human Gene Therapy | 1998

Aerosolization of Cationic Lipid:pDNA Complexes—In Vitro Optimization of Nebulizer Parameters for Human Clinical Studies

Simon J. Eastman; Jennifer D. Tousignant; Michael Lukason; Qiuming Chu; Seng H. Cheng; Ronald K. Scheule


Molecular Therapy | 2005

Local Delivery of a Viral Vector Mitigates Neutralization by Antiviral Antibodies and Results in Efficient Transduction of Rabbit Liver

Bradley L. Hodges; Kristin M. Taylor; Qiuming Chu; Samantha E. Scull; Rebecca G. Serriello; Scott C. Anderson; Fei Wang; Ronald K. Scheule


Archive | 1999

CATIONIC AMPHIPHILE MICELLAR COMPLEXES

Jennifer D. Tousignant; Simon J. Eastman; Edward R. Lee; Ronald K. Scheule; Seng H. Cheng; Jennifer B. Nietupski; Qiuming Chu; John Marshall


Molecular Therapy | 2005

Transient siRNA-Mediated Attenuation of Liver Expression from an α-Galactosidase a Plasmid Reduces Subsequent Humoral Immune Responses to the Transgene Product in Mice

Qiuming Chu; Macy Joseph; Malgorzata Przybylska; Nelson S. Yew; Ronald K. Scheule

Collaboration


Dive into the Qiuming Chu's collaboration.

Researchain Logo
Decentralizing Knowledge