RT Journal Article SR Electronic T1 Reprogramming human T cell function and specificity with non-viral genome targeting JF bioRxiv FD Cold Spring Harbor Laboratory SP 183418 DO 10.1101/183418 A1 Theodore L. Roth A1 Cristina Puig-Saus A1 Ruby Yu A1 Eric Shifrut A1 Julia Carnevale A1 Joseph Hiatt A1 Justin Saco A1 Han Li A1 Jonathan Li A1 Victoria Tobin A1 David Nguyen A1 Andrea M. Ferris A1 Jeff Chen A1 Jean-Nicolas Schickel A1 Laurence Pellerin A1 David Carmody A1 Gorka Alkorta-Aranburu A1 Daniela Del Gaudio A1 Hiroyuki Matsumoto A1 Montse Morell A1 Ying Mao A1 Rolen Quadros A1 Channabasavaiah Gurumurthy A1 Baz Smith A1 Michael Haugwitz A1 Stephen H. Hughes A1 Jonathan Weissman A1 Kathrin Schumann A1 Andrew P. May A1 Alan Ashworth A1 Gary Kupfer A1 Siri Greeley A1 Rosa Bacchetta A1 Eric Meffre A1 Maria Grazia Roncarolo A1 Neil Romberg A1 Kevan C. Herold A1 Antoni Ribas A1 Manuel D. Leonetti A1 Alexander Marson YR 2017 UL http://biorxiv.org/content/early/2017/12/07/183418.abstract AB Human T cells are central to physiological immune homeostasis, which protects us from pathogens without collateral autoimmune inflammation. They are also the main effectors in most current cancer immunotherapy strategies1. Several decades of work have aimed to genetically reprogram T cells for therapeutic purposes2–5, but as human T cells are resistant to most standard methods of large DNA insertion these approaches have relied on recombinant viral vectors, which do not target transgenes to specific genomic sites6, 7. In addition, the need for viral vectors has slowed down research and clinical use as their manufacturing and testing is lengthy and expensive. Genome editing brought the promise of specific and efficient insertion of large transgenes into target cells through homology-directed repair (HDR), but to date in human T cells this still requires viral transduction8, 9. Here, we developed a non-viral, CRISPR-Cas9 genome targeting system that permits the rapid and efficient insertion of individual or multiplexed large (>1 kilobase) DNA sequences at specific sites in the genomes of primary human T cells while preserving cell viability and function. We successfully tested the potential therapeutic use of this approach in two settings. First, we corrected a pathogenic IL2RA mutation in primary T cells from multiple family members with monogenic autoimmune disease and demonstrated enhanced signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR redirecting T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized the tumour antigen, with concomitant cytokine release and tumour cell killing. Taken together, these studies provide preclinical evidence that non-viral genome targeting will enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.