Rational plasmid design and bioprocess optimization to enhance recombinant adeno-associated virus (AAV) productivity in mammalian cells

Biotechnol J. 2016 Feb;11(2):290-7. doi: 10.1002/biot.201500176. Epub 2015 Sep 24.

Abstract

Viral vectors used for gene and oncolytic therapy belong to the most promising biological products for future therapeutics. Clinical success of recombinant adeno-associated virus (rAAV) based therapies raises considerable demand for viral vectors, which cannot be met by current manufacturing strategies. Addressing existing bottlenecks, we improved a plasmid system termed rep/cap split packaging and designed a minimal plasmid encoding adenoviral helper function. Plasmid modifications led to a 12-fold increase in rAAV vector titers compared to the widely used pDG standard system. Evaluation of different production approaches revealed superiority of processes based on anchorage- and serum-dependent HEK293T cells, exhibiting about 15-fold higher specific and volumetric productivity compared to well-established suspension cells cultivated in serum-free medium. As for most other viral vectors, classical stirred-tank bioreactor production is thus still not capable of providing drug product of sufficient amount. We show that manufacturing strategies employing classical surface-providing culture systems can be successfully transferred to the new fully-controlled, single-use bioreactor system Integrity(TM) iCELLis(TM) . In summary, we demonstrate substantial bioprocess optimizations leading to more efficient and scalable production processes suggesting a promising way for flexible large-scale rAAV manufacturing.

Keywords: Adeno-associated virus; HEK293T; Recombinant vector; Vector production; iCELLis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Bioengineering / methods*
  • Dependovirus / genetics
  • Dependovirus / physiology*
  • HEK293 Cells
  • HeLa Cells
  • Humans
  • Plasmids / genetics*
  • Virus Replication