Nature Communications (Aug 2024)

Systematic multi-trait AAV capsid engineering for efficient gene delivery

  • Fatma-Elzahraa Eid,
  • Albert T. Chen,
  • Ken Y. Chan,
  • Qin Huang,
  • Qingxia Zheng,
  • Isabelle G. Tobey,
  • Simon Pacouret,
  • Pamela P. Brauer,
  • Casey Keyes,
  • Megan Powell,
  • Jencilin Johnston,
  • Binhui Zhao,
  • Kasper Lage,
  • Alice F. Tarantal,
  • Yujia A. Chan,
  • Benjamin E. Deverman

DOI
https://doi.org/10.1038/s41467-024-50555-y
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 14

Abstract

Read online

Abstract Broadening gene therapy applications requires manufacturable vectors that efficiently transduce target cells in humans and preclinical models. Conventional selections of adeno-associated virus (AAV) capsid libraries are inefficient at searching the vast sequence space for the small fraction of vectors possessing multiple traits essential for clinical translation. Here, we present Fit4Function, a generalizable machine learning (ML) approach for systematically engineering multi-trait AAV capsids. By leveraging a capsid library that uniformly samples the manufacturable sequence space, reproducible screening data are generated to train accurate sequence-to-function models. Combining six models, we designed a multi-trait (liver-targeted, manufacturable) capsid library and validated 88% of library variants on all six predetermined criteria. Furthermore, the models, trained only on mouse in vivo and human in vitro Fit4Function data, accurately predicted AAV capsid variant biodistribution in macaque. Top candidates exhibited production yields comparable to AAV9, efficient murine liver transduction, up to 1000-fold greater human hepatocyte transduction, and increased enrichment relative to AAV9 in a screen for liver transduction in macaques. The Fit4Function strategy ultimately makes it possible to predict cross-species traits of peptide-modified AAV capsids and is a critical step toward assembling an ML atlas that predicts AAV capsid performance across dozens of traits.