Abstract

Of the 12 common serotypes used for gene delivery applications, Adeno-associated virus (AAV)rh.10 serotype has shown sustained hepatic transduction and has the lowest seropositivity in humans. We have evaluated if further modifications to AAVrh.10 at its phosphodegron like regions or predicted immunogenic epitopes could improve its hepatic gene transfer and immune evasion potential. Mutant AAVrh.10 vectors were generated by site directed mutagenesis of the predicted targets. These mutant vectors were first tested for their transduction efficiency in HeLa and HEK293T cells. The optimal vector was further evaluated for their cellular uptake, entry, and intracellular trafficking by quantitative PCR and time-lapse confocal microscopy. To evaluate their potential during hepatic gene therapy, C57BL/6 mice were administered with wild-type or optimal mutant AAVrh.10 and the luciferase transgene expression was documented by serial bioluminescence imaging at 14, 30, 45, and 72 days post-gene transfer. Their hepatic transduction was further verified by a quantitative PCR analysis of AAV copy number in the liver tissue. The optimal AAVrh.10 vector was further evaluated for their immune escape potential, in animals pre-immunized with human intravenous immunoglobulin. Our results demonstrate that a modified AAVrh.10 S671A vector had enhanced cellular entry (3.6 fold), migrate rapidly to the perinuclear region (1 vs. >2 h for wild type vectors) in vitro, which further translates to modest increase in hepatic gene transfer efficiency in vivo. More importantly, the mutant AAVrh.10 vector was able to partially evade neutralizing antibodies (~27–64 fold) in pre-immunized animals. The development of an AAV vector system that can escape the circulating neutralizing antibodies in the host will substantially widen the scope of gene therapy applications in humans.

Highlights

  • Gene therapy using recombinant Adeno-associated virus (Serotypes AAV1 to AAV12) has gained significant attention after early success with several pre-clinical and clinical studies (De et al, 2006; Zincarelli et al, 2008; Mendell et al, 2010; Mingozzi and High, 2011; Nathwani et al, 2011)

  • We have generated a total of nine mutant AAVrh.10 vectors by targeting particular S, T, K amino acids on the capsid that are homologous to the phosphodegrons reported in AAV2 capsid

  • The present study was designed to understand if modification of phosphodegron-like regions in an alternate serotype AAVrh.10 can improve (a) cellular uptake (b) intracellular trafficking (c) transgene expression and (d) minimize immune-recognition by circulating neutralizing antibodies (Nab) to facilitate gene transfer in a sero-positive setting

Read more

Summary

Introduction

Gene therapy using recombinant Adeno-associated virus (Serotypes AAV1 to AAV12) has gained significant attention after early success with several pre-clinical and clinical studies (De et al, 2006; Zincarelli et al, 2008; Mendell et al, 2010; Mingozzi and High, 2011; Nathwani et al, 2011). A similar immunogenic response to AAV is evident from other clinical trials as well (Mcphee et al, 2006; Brantly et al, 2009) These data suggest that any long term AAV vector transgene expression will require approaches to minimize capsid specific cellular immune responses. This will need AAV vectors that express therapeutic levels of the transgene at low vector doses to circumvent dose dependent immunotoxicity (Manno et al, 2006; Mingozzi et al, 2007)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call