-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

200 ZFN-Mediated Gene Targeting at the Albumin Locus in Liver Results in Therapeutic Levels of Human FIX in Mice and Non-Human Primates

Gene Therapy and Transfer
Program: Oral and Poster Abstracts
Type: Oral
Session: 801. Gene Therapy and Transfer: Gene Therapy for Hemoglobinopathies and Inherited Bleeding Disorders
Sunday, December 6, 2015: 7:45 AM
Tangerine 3 (WF3-4), Level 2 (Orange County Convention Center)

Thomas Wechsler, PhD1*, Kathleen E. Meyer2*, S. Kaye Spratt2*, Judith Greengard, PhD1*, Yolanda Santiago3*, Scott Sproul3*, Richard Surosky4*, David E. Paschon2*, Nathalie Dubois-Stringfellow2*, Dale Genji Ando, MD2*, Geoff Nichol5*, Edward J. Rebar, PhD1* and Michael C. Holmes, PhD1*

1Sangamo BioSciences, Richmond, CA
2Sangamo Biosciences Inc, Richmond, CA
3Sangamo Biosciences Inc., Richmond, CA
4Sangamo Biosciences Inc, Richmond, CA, Afghanistan
5Sangamo Biosciences Inc, RIchmond, CA

Hemophilia is an attractive target for gene therapy, since activity levels as low as 1% to 2% of normal are beneficial and levels of ~5% prevent spontaneous bleeding. Our goal was to provide a single treatment that permanently enables hepatic production of therapeutic levels of hFIX activity to decrease or potentially eliminate the need for prophylactic treatment in hemophilia B patients. We performed targeted in vivo genome editing using 1) two zinc finger nucleases (ZFNs) targeting intron 1 of the albumin locus, and 2) a human F9 donor template construct. The ZFNs and donor template are encoded on separate hepatotropic adeno-associated virus serotype 2/6 (AAV2/6) vectors injected intravenously, resulting in targeted insertion of a corrected copy of the hF9 gene into the albumin locus in a proportion of liver hepatocytes. The albumin locus was selected as a “safe harbor” as production of this most abundant plasma protein exceeds 10 g/day, and moderate reductions in those levels are well-tolerated.  These genome edited hepatocytes produce normal hFIX in therapeutic quantities, rather than albumin, driven by the highly active albumin enhancer/promoter, to treat hemophilia B; the genetic modification is expected to be sustained even in the face of hepatocyte turnover, making this approach attractive for treating young children with hemophilia before the appearance of significant organ damage.

Transformed and primary human hepatocytes transduced in vitro with AAV2/6 encoding human albumin ZFNs and a promoterless hF9 transgene were shown to secrete hFIX. Extensive molecular analyses demonstrated that this was due to targeted integration of the hF9 transgene at the albumin locus and splicing of this gene into the albumin transcript. By employing AAV2/6 delivery of murine-specific ZFNs in vivo, stable levels of hFIX were observed in blood of mice injected with the albumin ZFNs and hF9 transgene donor. C57BL/6 mice were administered vehicle (n=20) or AAV2/6 vectors (n=25) encoding mouse surrogate reagents at 1.0 x1013 vector genome (vg)/kg via tail vein injection. ELISA analysis of plasma hFIX in the treated mice showed peak levels of 50-1053 ng/mL that were sustained for the duration of the 6-month study. Analysis of FIX activity from mouse plasma confirmed bioactivity commensurate with expression levels. Next, we report the feasibility of this approach in non-human primates (NHPs), showing that a single intravenous co-infusion of AAV2/6 vectors encoding the NHP targeted albumin-specific ZFNs and a human F9 donor at 1.2x1013 vg/kg (n=5/group) resulted in >50 ng/mL (>1% of normal) in this large animal model. The use of higher AAV2/6 doses (up to 1.5x1014 vg/kg) yielded plasma hFIX levels up to 1000 ng/ml (or 20% of normal) in several animals and up to 2000 ng/ml (or 50% of normal) in a single animal, for the duration of the study (3 months). The treatment was well tolerated in mice and NHPs, with no significant toxicological findings related to AAV2/6 ZFN + donor treatment in either species at therapeutic doses. Together, these data support a clinical trial to determine if a single co-administration of ZFN and donor AAV vectors is sufficient to enable therapeutic and potentially lifelong production of the clotting factor for the treatment of Hemophilia B.

Disclosures: Wechsler: Sangamo BioSciences: Employment . Meyer: Sangamo Biosciences Inc: Employment . Spratt: Sangamo Biosciences Inc: Employment . Greengard: Sangamo Biosciences Inc: Employment . Santiago: Sangamo Biosciences Inc: Employment . Sproul: Sangamo Biosciences Inc: Employment . Surosky: Sangamo Biosciences Inc: Employment . Paschon: Sangamo Biosciences Inc: Employment . Dubois-Stringfellow: Sangamo Biosciences Inc: Employment . Ando: Sangamo Biosciences Inc: Employment . Nichol: Sangamo Biosciences Inc: Employment . Rebar: Sangamo BioSciences: Employment . Holmes: Sangamo BioSciences: Employment .

*signifies non-member of ASH