4.8 Article

Next generation of adeno-associated virus 2 vectors: Point mutations in tyrosines lead to high-efficiency transduction at lower doses

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.0802866105

Keywords

aAV vectors; capsid mutations; gene expression; gene therapy; gene transfer

Funding

  1. NHLBI NIH HHS [P01 HL-51811, P01 HL051811, R01 HL-65570, P01 HL078810, P01 HL059412, R01 HL-07691, R01 HL065570, P01 HL-078810] Funding Source: Medline
  2. NIAID NIH HHS [R01 AI051390] Funding Source: Medline
  3. NIBIB NIH HHS [R01 EB-002073, R01 EB002073] Funding Source: Medline
  4. NIDDK NIH HHS [R01 DK062302, R01 DK-062302, P01 DK 058327, P01 DK058327] Funding Source: Medline
  5. NIGMS NIH HHS [R01 GM082946, R01 GM-082946] Funding Source: Medline

Ask authors/readers for more resources

Recombinant adeno-associated virus 2 (AAV2) vectors are in use in several Phase I/II clinical trials, but relatively large vector doses are needed to achieve therapeutic benefits. Large vector doses also trigger an immune response as a significant fraction of the vectors fails to traffic efficiently to the nucleus and is targeted for degradation by the host cell proteasome machinery. We have reported that epidermal growth factor receptor protein tyrosine kinase (EGFR-PTK) signaling negatively affects transduction by AAV2 vectors by impairing, nuclear transport of the vectors. We have also observed that EGFR-PTK can phosphorylate AAV2 capsids at tyrosine residues. Tyrosine-phosphorylated AAV2 vectors enter cells efficiently but fail to transduce effectively, in part because of ubiquitination of AAV capsids followed by proteasome-mediated degradation. We reasoned that mutations of the surface-exposed tyrosine residues might allow the vectors to evade phosphorylation and subsequent ubiquitination and, thus, prevent proteasome-mediated degradation. Here, we document that site-directed mutagenesis of surface-exposed tyrosine residues leads to production of vectors that transduce HeLa cells approximate to 10-fold more efficiently in vitro and murine hepatocytes nearly 30-fold more efficiently in vivo at a log lower vector dose. Therapeutic levels of human Factor IX (FAX) are also produced at an approximate to 10-fold reduced vector dose. The increased transduction efficiency of tyrosine-mutant vectors is due to lack of capsid ubiquitination and improved intracellular trafficking to the nucleus. These studies have led to the development of AAV vectors that are capable of high-efficiency transduction at lower doses, which has important implications in their use in human gene therapy.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available