Network


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

Hotspot


Dive into the research topics where Helen G Franck is active.

Publication


Featured researches published by Helen G Franck.


Blood | 2012

Prolonged activity of a recombinant factor VIII-Fc fusion protein in hemophilia A mice and dogs

Jennifer A. Dumont; Tongyao Liu; Susan C. Low; Xin Zhang; George D. Kamphaus; Paul Sakorafas; Cara Fraley; Douglas Drager; Thomas Reidy; Justin McCue; Helen G Franck; Elizabeth P. Merricks; Timothy C. Nichols; Alan J. Bitonti; Glenn F. Pierce; Haiyan Jiang

Despite proven benefits, prophylactic treatment for hemophilia A is hampered by the short half-life of factor VIII. A recombinant factor VIII-Fc fusion protein (rFVIIIFc) was constructed to determine the potential for reduced frequency of dosing. rFVIIIFc has an ∼ 2-fold longer half-life than rFVIII in hemophilia A (HemA) mice and dogs. The extension of rFVIIIFc half-life requires interaction of Fc with the neonatal Fc receptor (FcRn). In FcRn knockout mice, the extension of rFVIIIFc half-life is abrogated, and is restored in human FcRn transgenic mice. The Fc fusion has no impact on FVIII-specific activity. rFVIIIFc has comparable acute efficacy as rFVIII in treating tail clip injury in HemA mice, and fully corrects whole blood clotting time (WBCT) in HemA dogs immediately after dosing. Furthermore, consistent with prolonged half-life, rFVIIIFc shows 2-fold longer prophylactic efficacy in protecting HemA mice from tail vein transection bleeding induced 24-48 hours after dosing. In HemA dogs, rFVIIIFc also sustains partial correction of WBCT 1.5- to 2-fold longer than rFVIII. rFVIIIFc was well tolerated in both species. Thus, the rescue of FVIII by Fc fusion to provide prolonged protection presents a novel pathway for FVIII catabolism, and warrants further investigation.


Blood | 2010

Eradication of neutralizing antibodies to factor VIII in canine hemophilia A after liver gene therapy

Jonathan D. Finn; Margareth Castro Ozelo; Denise E. Sabatino; Helen G Franck; Elizabeth P. Merricks; Julie M. Crudele; Shangzhen Zhou; Haig H. Kazazian; David Lillicrap; Timothy C. Nichols; Valder R. Arruda

Inhibitory antibodies to factor VIII (FVIII) are a major complication in the treatment of hemophilia A, affecting approximately 20% to 30% of patients. Current treatment for inhibitors is based on long-term, daily injections of large amounts of FVIII protein. Liver-directed gene therapy has been used to induce antigen-specific tolerance, but there are no data in hemophilic animals with pre-existing inhibitors. To determine whether sustained endogenous expression of FVIII could eradicate inhibitors, we injected adeno-associated viral vectors encoding canine FVIII (cFVIII) in 2 strains of inhibitor hemophilia A dogs. In 3 dogs, a transient increase in inhibitor titers (up to 7 Bethesda Units [BU]) at 2 weeks was followed by continuous decline to complete disappearance within 4-5 weeks. Subsequently, an increase in cFVIII levels (1.5%-8%), a shortening of clotting times, and a reduction (> 90%) of bleeding episodes were observed. Immune tolerance was confirmed by lack of antibody formation after repeated challenges with cFVIII protein and normal protein half-life. A fourth dog exhibited a strong early anamnestic response (216 BU), with slow decline to 0.8 BU and cFVIII antigen detection by 18 months after vector delivery. These data suggest that liver gene therapy has the potential to eradicate inhibitors and could improve the outcomes of hemophilia A patients.


Blood | 2010

Peripheral transvenular delivery of adeno-associated viral vectors to skeletal muscle as a novel therapy for hemophilia B

Valder R. Arruda; Hansell H. Stedman; Virginia Haurigot; George Buchlis; Stefano Baila; Patricia Favaro; Yifeng Chen; Helen G Franck; Shangzhen Zhou; J. Fraser Wright; Linda B. Couto; Haiyan Jiang; Glenn F. Pierce; Dwight A. Bellinger; Federico Mingozzi; Timothy C. Nichols; Katherine A. High

Muscle represents an important tissue target for adeno-associated viral (AAV) vector-mediated gene transfer of the factor IX (FIX) gene in hemophilia B (HB) subjects with advanced liver disease. Previous studies of direct intramuscular administration of an AAV-FIX vector in humans showed limited efficacy. Here we adapted an intravascular delivery system of AAV vectors encoding the FIX transgene to skeletal muscle of HB dogs. The procedure, performed under transient immunosuppression (IS), resulted in widespread transduction of muscle and sustained, dose-dependent therapeutic levels of canine FIX transgene up to 10-fold higher than those obtained by intramuscular delivery. Correction of bleeding time correlated clinically with a dramatic reduction of spontaneous bleeding episodes. None of the dogs (n = 14) receiving the AAV vector under transient IS developed inhibitory antibodies to canine FIX; transient inhibitor was detected after vector delivery without IS. The use of AAV serotypes with high tropism for muscle and low susceptibility to anti-AAV2 antibodies allowed for efficient vector administration in naive dogs and in the presence of low- but not high-titer anti-AAV2 antibodies. Collectively, these results demonstrate the feasibility of this approach for treatment of HB and highlight the importance of IS to prevent immune responses to the FIX transgene product.


Molecular Therapy | 2011

Efficacy and Safety of Long-term Prophylaxis in Severe Hemophilia A Dogs Following Liver Gene Therapy Using AAV Vectors

Denise E. Sabatino; Amy M. Lange; Ekaterina Altynova; Rita Sarkar; Shangzhen Zhou; Elizabeth P. Merricks; Helen G Franck; Timothy C. Nichols; Valder R. Arruda; Haig H. Kazazian

Developing adeno-associated viral (AAV)-mediated gene therapy for hemophilia A (HA) has been challenging due to the large size of the factor VIII (FVIII) complementary DNA and the concern for the development of inhibitory antibodies to FVIII in HA patients. Here, we perform a systematic study in HA dogs by delivering a canine FVIII (cFVIII) transgene either as a single chain or two chains in an AAV vector. An optimized cFVIII single chain delivered using AAV serotype 8 (AAV8) by peripheral vein injection resulted in a dose-response with sustained expression of FVIII up to 7% (n = 4). Five HA dogs administered two-chain delivery using either AAV8 or AAV9 via the portal vein expressed long-term, vector dose-dependent levels of FVIII activity (up to 10%). In the two-chain approach, circulating cFVIII antigen levels were more than fivefold higher than activity. Notably, no long-term immune response to FVIII was observed in any of the dogs (1/9 dogs had a transient inhibitor). Long-term follow-up of the dogs showed a remarkable reduction (>90%) of bleeding episodes in a combined total of 24 years of observation. These data demonstrate that both approaches are safe and achieve dose-dependent therapeutic levels of FVIII expression, which supports translational studies of AAV-mediated delivery for HA.


Molecular Therapy | 2010

Hyperactive sleeping beauty transposase enables persistent phenotypic correction in mice and a canine model for hemophilia B.

Martin Hausl; Wenli Zhang; Nadine Müther; Christina Rauschhuber; Helen G Franck; Elizabeth P. Merricks; Timothy C. Nichols; Mark A. Kay; Anja Ehrhardt

Sleeping Beauty (SB) transposase enables somatic integration of exogenous DNA in mammalian cells, but potency as a gene transfer vector especially in large mammals has been lacking. Herein, we show that hyperactive transposase system delivered by high-capacity adenoviral vectors (HC-AdVs) can result in somatic integration of a canine factor IX (cFIX) expression-cassette in canine liver, facilitating stabilized transgene expression and persistent haemostatic correction of canine hemophilia B with negligible toxicity. We observed stabilized cFIX expression levels during rapid cell cycling in mice and phenotypic correction of the bleeding diathesis in hemophilia B dogs for up to 960 days. In contrast, systemic administration of an inactive transposase system resulted in rapid loss of transgene expression and transient phenotypic correction. Notably, in dogs a higher viral dose of the active SB transposase system resulted into transient phenotypic correction accompanied by transient increase of liver enzymes. Molecular analysis of liver samples revealed SB-mediated integration and provide evidence that transgene expression was derived mainly from integrated vector forms. Demonstrating that a viral vector system can deliver clinically relevant levels of a therapeutic protein in a large animal model of human disease paves a new path toward the possible cure of genetic diseases.


Molecular Therapy | 2010

Safety of AAV Factor IX Peripheral Transvenular Gene Delivery to Muscle in Hemophilia B Dogs

Virginia Haurigot; Federico Mingozzi; George Buchlis; Daniel J. Hui; Yifeng Chen; Etiena Basner-Tschakarjan; Valder R. Arruda; Antoneta Radu; Helen G Franck; J. Fraser Wright; Shangzhen Zhou; Hansell H. Stedman; Dwight A. Bellinger; Timothy C. Nichols; Katherine A. High

Muscle represents an attractive target tissue for adeno-associated virus (AAV) vector–mediated gene transfer for hemophilia B (HB). Experience with direct intramuscular (i.m.) administration of AAV vectors in humans showed that the approach is safe but fails to achieve therapeutic efficacy. Here, we present a careful evaluation of the safety profile (vector, transgene, and administration procedure) of peripheral transvenular administration of AAV-canine factor IX (cFIX) vectors to the muscle of HB dogs. Vector administration resulted in sustained therapeutic levels of cFIX expression. Although all animals developed a robust antibody response to the AAV capsid, no T-cell responses to the capsid antigen were detected by interferon (IFN)-γ enzyme-linked immunosorbent spot (ELISpot). Interleukin (IL)-10 ELISpot screening of lymphocytes showed reactivity to cFIX-derived peptides, and restimulation of T cells in vitro in the presence of the identified cFIX epitopes resulted in the expansion of CD4+FoxP3+IL-10+ T-cells. Vector administration was not associated with systemic inflammation, and vector spread to nontarget tissues was minimal. At the local level, limited levels of cell infiltrates were detected when the vector was administered intravascularly. In summary, this study in a large animal model of HB demonstrates that therapeutic levels of gene transfer can be safely achieved using a novel route of intravascular gene transfer to muscle.


Haemophilia | 2010

Prevention of spontaneous bleeding in dogs with haemophilia A and haemophilia B

Timothy C. Nichols; Robin A. Raymer; Helen G Franck; Elizabeth P. Merricks; Dwight A. Bellinger; N. Defriess; Paris Margaritis; Valder R. Arruda; Mark A. Kay; Katherine A. High

Summary.  Dogs with haemophilia A or haemophilia B exhibit spontaneous bleeding comparable with the spontaneous bleeding phenotype that occurs in humans with severe haemophilia. The phenotypic and genotypic characteristics of haemophilic dogs have been well‐described, and such dogs are suitable for testing prophylactic protein replacement therapy and gene transfer strategies. In dogs with haemophilia, long‐term effects on spontaneous bleeding frequency (measured over years) can be used as an efficacy endpoint in such studies. Although complete correction of coagulopathy has not been achieved, published data show that prophylactic factor replacement therapy and gene transfer can markedly reduce the frequency of spontaneous bleeding in haemophilic dogs. Further studies are currently ongoing.


Ilar Journal | 2009

Protein replacement therapy and gene transfer in canine models of hemophilia A, hemophilia B, von Willebrand disease, and factor VII deficiency

Timothy C. Nichols; Aaron Dillow; Helen G Franck; Elizabeth P. Merricks; Robin A. Raymer; Dwight A. Bellinger; Valder R. Arruda; Katherine A. High


Blood | 2009

Factor VIII-Fc Fusion Protein Shows Extended Half-Life and Hemostatic Activity in Hemophilia A Dogs.

Jennifer A. Dumont; George D. Kamphaus; Cara Fraley; Tamera Ashworth; Helen G Franck; Elizabeth P. Merricks; Timothy C. Nichols; Alan J. Bitonti


Blood | 2008

Recombinant Interleukin-11 (rhIL-11) in Women with Refractory Menorrhagia and Von Willebrand Disease.

Rachel C. Jankowitz; Margaret V. Ragni; Holly L. Chapman; Elizabeth P. Merricks; Mark T. Kloos; Aaron Dillow; Helen G Franck; Timothy C. Nichols

Collaboration


Dive into the Helen G Franck's collaboration.

Top Co-Authors

Avatar

Timothy C. Nichols

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Elizabeth P. Merricks

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Valder R. Arruda

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Dwight A. Bellinger

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Katherine A. High

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Shangzhen Zhou

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Aaron Dillow

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

David A. Wilcox

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Denise E. Sabatino

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Eric S. Jensen

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge