scholarly journals Gene transfer to hemophilia A mice via oral delivery of FVIII–chitosan nanoparticles

2008 ◽  
Vol 132 (3) ◽  
pp. 252-259 ◽  
Author(s):  
Katherine Bowman ◽  
Rita Sarkar ◽  
Sanj Raut ◽  
Kam W. Leong
1997 ◽  
Vol 46 (1-2) ◽  
pp. 75-87 ◽  
Author(s):  
Elke Walter ◽  
Maria A Croyle ◽  
Beverly L Davidson ◽  
Blake J Roessler ◽  
John M Hilfinger ◽  
...  

Blood ◽  
2009 ◽  
Vol 113 (16) ◽  
pp. 3682-3689 ◽  
Author(s):  
Paris Margaritis ◽  
Elise Roy ◽  
Majed N. Aljamali ◽  
Harre D. Downey ◽  
Urs Giger ◽  
...  

Abstract Continuous expression of activated factor VII (FVIIa) via gene transfer is a potential therapeutic approach for hemophilia patients with or without inhibitory antibodies to human factor VIII (FVIII) or IX (FIX). Here, we investigate whether gene transfer of an engineered canine FVIIa (cFVIIa) transgene can affect hemostasis in a canine model of hemophilia, a good predictor of efficacy of hemophilia treatments. Purified recombinant cFVIIa exhibited 12-fold higher tissue factor–dependent activity than purified recombinant zymogen cFVII. Subsequently, we generated a serotype 8 recombinant adeno-associated viral vector expressing cFVIIa from a liver-specific promoter. Vector delivery via the portal vein in hemophilia A and B dogs was well tolerated, and long-term expression of cFVIIa resulted in a shortening of the prothrombin time, partial correction of the whole blood clotting time and thromboelastography parameters, and a complete absence of spontaneous bleeding episodes. No evidence of hepatotoxicity, thrombotic complications, or inhibitory immune response was found. These data provide the first evidence for in vivo efficacy and safety of continuously expressed FVIIa as a FVIII/FIX-bypassing agent in a large animal model of hemophilia, avoiding the risk of inhibitor formation associated with bolus FVIII or FIX infusion.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3238-3238
Author(s):  
Irene Zolotukhin ◽  
Brett Palaschak ◽  
David M Markusic ◽  
Roland Herzog

Abstract Hemophilia A is the X-linked bleeding disorder resulting from the loss of functional clotting factor VIII (FVIII). Hemophilia A patients with severe disease (<1% residual FVIII activity) experience spontaneous bleeds into the joints and closed spaces with severe morbidity. Restoration of hemostasis is managed by repeated infusions (2-3 times per week) of plasma derived or recombinant FVIII protein. While a standard treatment is available for patients, life-long infusions of FVIII protein is very expensive, has a negative impact on the patient's quality of life, and FVIII protein products are not available worldwide. Hence, there is a need to develop a more robust and cost effective treatment for hemophilia A patients. Liver-directed gene therapy using adeno-associated virus (AAV) represents a promising approach to treat hemophilia A. However, previous studies have shown that overexpression of human FVIII protein in the context of hydrodynamic delivery of plasmid vectors induces ER stress mediated through the unfolded protein response (UPR). Because human FVIII protein is inefficiently secreted into circulation, high AAV vector doses will be required to obtain therapeutic expression levels. Therefore, we sought to determine if AAV-FVIII gene delivery also triggers cellular UPR in hepatocytes in vivo. To this end, we selected to use a codon-optimized FVIII cDNA, which has been shown by our lab to significantly increase FVIII protein expression, and a high vector dose of AAV8-ApoE-hAAT-cohF8, containing a hepatocyte-specific enhancer/promoter combination. We evaluated this vector at doses of 1 x 1011 vg (4x1012 vg/kg) and 1 x 1012 vg (4x1013 vg/kg) in hemophilia A mice on a 129/BL6 background. Intravenous administration of the highest vector dose completely restored hemostasis, which was sustained and achieved super-physiological levels in some animals. Importantly, none of these mice developed inhibitors against FVIII. Next, we administered the vector at the same 2 doses to C67BL/6 mice, which show higher hepatic transduction efficiency than other strains. Experimental controls consisted of mice, with no vector or 1x1012 vg of AAV8-ApoE-hAAT-F9, expressing human factor IX (FIX) protein. Injection of tunicamycin, a potent inducer of the ER stress response, served as a positive control for all assays. Vector-treated mice were studied 2 and 4 weeks after gene transfer (n=3-4 per group). First, we evaluated the status of key molecular chaperones, known to be the mediators of the UPR: Bip, p-PERK, and p-eIF2a. Western blotting performed on the liver lysates indicated modest up-regulation of all three markers compared to normal control, but that effect was neither dose nor gene dependent. In addition, we tested the splicing of Xbp1 mRNA by PCR assay and observed low level of the 26 bp spliced fragment, indicative of the UPR, at the high vector dose. Immunohistochemistry on liver sections from each of our experimental groups including H&E staining, Tunnel staining for apoptotic cells, and reactive oxygen species staining. None of the stains yielded evidence for liver damage even in the 1x1012 AAV8-cohF8 treated mice compared to untreated controls. There was also no elevation of liver enzyme levels in plasma samples. Analysis of plasma from vector injected mice showed systemic levels of human FVIII and FIX proteins at ~30 ng/ml and ~6300 ng/ml, respectively (these ELISA-based measurements likely underestimate FVIII levels due to interference by von Willebrand factor). Overall our results suggest that over-expression of coagulation factors in hepatocytes from AAV vectors causes a mild cell stress response that is not strong enough to cause liver toxicity, is not specific for FVIII, and does impact expression or immunogenicity. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4415-4415 ◽  
Author(s):  
Reginald Tran ◽  
David R Myers ◽  
Jordan E Shields ◽  
Byungwook Ahn ◽  
Yongzhi Qiu ◽  
...  

Abstract Background: Recent clinical trials have demonstrated the efficacy and safety of gene therapy utilizing HIV-derived lentiviral vectors (LVs) for blood disorders. However, the LV requirements and clinical ex vivo cell transduction protocols used in these studies exposes the limitations of the technology and beckons the need for improved LV manufacturing and clinical transduction efficiency. Many methods have been devised to enhance efficiency, although none have circumvented the exorbitant amounts of virus required to achieve therapeutic HSC transduction. Furthermore, prolonged ex vivo cell culture is necessary to achieve sufficient transduction despite exposure to toxic byproducts of LV production. To that end, we developed a novel, scalable microfluidic for clinical LV transduction that leverages mass transfer principles to significantly reduce the amount of LV required to achieve therapeutic levels of gene transfer and transduction time by more efficiently exposing cells to virus. Results: Jurkats were transduced with a GFP-encoding clinical LV in microfluidics with surface areas (SAs) comparable to the bottom surface of 96 and 6-well plates. Microfluidic transductions were compared to well plate transductions with matched SA, cell numbers, viral particles, and incubation times. After LV incubation, cells were removed from the microfluidics and well plates, spun down, re-suspended with fresh media, and cultured for at least 72 hours at 37°C and 5% CO2. Cells were assessed for GFP expression with flow cytometry. Preliminary mouse studies utilized Sca+ cells isolated from CD45.1 donor mice via positive selection. The cells were transduced in the scaled up microfluidic with a bioengineered coagulation factor VIII (fVIII) transgene encoding LV and transplanted into host hemophilia A mice after myeloablative conditioning. Two weeks post-transplantation, blood samples were taken from the recipient animals and assayed for donor cell engraftment by flow cytometry and plasma fVIII activity by chromogenic assay. The high SA:volume ratio of the microfluidic enhances transduction by physically bringing cells and virus into closer proximity and enabling high concentrations of virus to be used without increasing the amount of virus set by the minimum volume requirements of LV transduction platforms (Fig. 1A). The polystyrene bottom of the microfluidic allows for Retronectin coating that immobilizes non-adherent cells on the bottom surface. LV can then be perfused at low concentrations to maintain a constant supply of fresh virus to the cells, increase convective mixing, and to minimize cell exposure to the toxic byproducts of LV production (Fig. 1B). These microfluidics have been scaled up to accommodate 106 cells, with potential to scale up to 107-108 cells (Fig. 1C). Cells transduced in the microfluidics showed 2-6 fold increases in GFP expression over well plates utilizing the same amount of cells, virus, and incubation times (Fig. 2A). The kinetics of LV transduction in the microfluidics also are faster, as seen by the steeper transduction curve. Five hours of incubation in the microfluidic yielded comparable transduction to 24 hours in the 6-well plate (Fig. 2B). Improvements in transduction also were observed by perfusing virus despite using lower virus concentrations (Fig. 2C). Finally, hemophilia A mice transplanted with donor CD45.1 Sca+ cells transduced in the microfluidic have engrafted (Fig. 3A) and produce fVIII (Fig. 3B) after two weeks with similar profiles to control cells transduced in a 6-well plate despite using half the amount of virus and shorter incubation times. Conclusions and ongoing efforts: We describe a novel microfluidic that significantly reduces the amount of virus and ex vivo processing time required for therapeutic levels of transduction in clinical gene therapy. This device is versatile in its compatibility with current transduction strategies such as Retronectin and polybrene in addition to offering new approaches to boosting gene transfer efficiency. Furthermore, we have shown that the device has clinical potential by successfully scaling up cell numbers and transplanting mice with microfluidic transduced cells, of which there is an ongoing effort to monitor fVIII production and determine virus copy number. Future work will involve optimization with transduction-enhancing compounds, further scaling, and continued in vivo experiments. Disclosures Spencer: Expression Therapeutics: Equity Ownership. Doering:Bayer Healthcare: Consultancy, Honoraria, Research Funding; Expression Therapeutics: Equity Ownership.


2006 ◽  
Vol 13 ◽  
pp. S33 ◽  
Author(s):  
Benjamin R. Harmeling ◽  
Steven Ziegler ◽  
Troy Torgerson ◽  
Liping Chen ◽  
Hans D. Ochs ◽  
...  

2014 ◽  
Vol 5 (11) ◽  
pp. 1191-1201 ◽  
Author(s):  
Sara M Navarro ◽  
Caleb Darensbourg ◽  
Linda Cross ◽  
Rhett Stout ◽  
Diana Coulon ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document