scholarly journals 546. Development and Characterization of a New Tet-ON AAV Vector for Tightly Controlled Transgene Expression

2011 ◽  
Vol 19 ◽  
pp. S210
2016 ◽  
Vol 24 ◽  
pp. S4-S5
Author(s):  
Claire A. Schreiber ◽  
Yoshihiro Izumiya ◽  
Aravind Asokan ◽  
Yasuhiro Ikeda

2008 ◽  
Vol 18 (3) ◽  
pp. 377-385 ◽  
Author(s):  
Jiedao Zhang ◽  
Longtao Lu ◽  
Lusha Ji ◽  
Guodong Yang ◽  
Chengchao Zheng

1997 ◽  
Vol 8 (1) ◽  
pp. 45-56 ◽  
Author(s):  
Johanne M. Kaplan ◽  
Donna Armentano ◽  
Tim E. Sparer ◽  
Susan G. Wynn ◽  
Pamela A. Peterson ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 751-751 ◽  
Author(s):  
Xavier M Anguela ◽  
Rajiv Sharma ◽  
Yannick Doyon ◽  
Sunnie Y Wong ◽  
David E Paschon ◽  
...  

Abstract Abstract 751 Gene correction using zinc finger nuclease (ZFN) technology can be applied to target virtually any locus in the human genome. Beyond correcting mutated genes causative of disease, ZFNs can also be utilized to target transgene insertion into genomic “safe harbors.” Ideally, specific gene targeting to such “safe harbor” sites would (i) ensure therapeutically relevant levels of transgene expression and (ii) tolerate transgene addition without deleterious effect on the host organism. For liver-derived protein replacement, albumin represents an attractive target locus. Firstly, albumin is very highly expressed exclusively in the liver, thus targeting of a relatively small percentage of alleles should yield therapeutically relevant levels of liver-specific transgene expression. Second, the reduction or complete absence of albumin in animals and even humans (analbuminemia) produces surprisingly few symptoms. Here, we sought to investigate whether ZFN-mediated targeted insertion of a promoter-less copy of the human F9 cDNA at the mouse albumin locus could result in human Factor IX production and successfully correct the hemophilic phenotype in mice. To address this question, we constructed an AAV vector encoding a pair of ZFNs targeting intron 1 of the mouse albumin locus (AAV8-mAlb-ZFN) and a donor AAV vector (AAV8-Donor) harboring a partial cDNA cassette containing exons 2–8 of the wild-type human F9 gene flanked by sequences lacking significant homology to the mouse genome. Co-delivery of 1e11 vg of AAV8-mAlb-ZFN along with 5e11vg of AAV8-Donor resulted in stable (>12wk) circulating F.IX levels of 1600–3200 ng/mL (32–64% of normal). As a control, mice injected with the AAV8-Donor along with an AAV vector encoding a ZFN pair targeting an unrelated locus exhibited background F.IX levels (∼50 ng/mL). A dose-response study was performed by administering a fixed dose of donor (5e11 vg/mouse) with decreasing doses of AAV8-mAlb-ZFN (1e11, 1e10 and 1e9 vg/mouse). Human F.IX levels increased as a function of ZFN dose in the range tested (3260±480, 225±43 and 31±4 ng/mL at the high, medium and low dose, respectively). Importantly, these results showed that donor homology to the target site is not required to achieve robust levels of gene addition to the albumin locus in adult mice, thus permitting the design of donor vectors harboring corrective copies of transgenes up to the maximum AAV packaging capacity of ∼4.7 Kb. Albumin and factor IX are both synthesized as pre-propeptides and turned into propeptides after the signal peptide is removed. Expression of human F9 exons 2–8 spliced with mouse albumin exon 1 is expected to yield a chimeric propeptide. The first 2 N-terminal amino acids would originate from proalbumin, followed by a Val to Leu mutation at position −17 of the hF.IX propeptide and 16 aa encoded by human F9. To evaluate whether this chimeric human F.IX derived from gene addition to the albumin locus would be processed correctly and normalize the prolonged clotting times in hemophilia B (HB) mice, we injected 1e11 vg of AAV8-mAlb-ZFN and 5e11vg of AAV8-Donor into HB animals. Two weeks post-treatment, hF.IX antigen levels were in the range of 20% of normal and activated partial thromboplastin time, a measurement of clot formation, was corrected to wild-type levels (42 seconds), from an average of 70 seconds pre-treatment. Thus expression of a therapeutic protein (F.IX) from the albumin locus is shown to correct the HB disease phenotype in vivo. In summary, these data provide the first demonstration of ZFN-mediated in vivo genome editing of a safe harbor locus for therapeutic protein production. While we provide here a proof of principle establishing phenotypic correction of hemophilia B, appropriately designed donors could expand this strategy. Most importantly the magnitude of albumin expression (>15 g / day) should enable production of a diverse range of transgenes at therapeutically consequential levels. Disclosures: Anguela: The Children's Hospital of Philadelphia: Patents & Royalties. Sharma:The Children's Hospital of Philadelphia: Patents & Royalties. Doyon:Sangamo BioSciences, Inc.: Employment. Wong:Sangamo BioSciences, Inc.: Employment. Paschon:Sangamo BioSciences, Inc.: Employment. Gregory:Sangamo BioSciences, Inc.: Employment. Holmes:Sangamo BioSciences, Inc.: Employment. Rebar:Sangamo BioSciences, Inc.: Employment. High:Shire Pharmaceuticals: Consultancy; Sangamo Biosciences, Inc: Collaborator, Collaborator Other; Novo Nordisk: Visiting Professor, Visiting Professor Other; Genzyme, Inc: Membership on an entity's Board of Directors or advisory committees; The Children's Hospital of Philadelphia: Patents & Royalties; Bluebird Bio, Inc: Membership on an entity's Board of Directors or advisory committees.


2006 ◽  
Vol 13 (6) ◽  
pp. 514-521 ◽  
Author(s):  
James W. Godwin ◽  
Nella Fisicaro ◽  
Anthony J. F. d'Apice ◽  
Peter J. Cowan

2015 ◽  
Vol 523 (10) ◽  
pp. 1474-1487 ◽  
Author(s):  
Cynthia L. Chow ◽  
Weixiang Guo ◽  
Parul Trivedi ◽  
Xinyu Zhao ◽  
Samuel P. Gubbels

2018 ◽  
Vol 44 (2) ◽  
pp. 181-194 ◽  
Author(s):  
Julien Bernaud ◽  
Axel Rossi ◽  
Anny Fis ◽  
Lara Gardette ◽  
Ludovic Aillot ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document