Reprogramming human T cell function and specificity with non-viral genome targeting

Decades of work have aimed to genetically reprogram T cells for therapeutic purposes1,2 using recombinant viral vectors, which do not target transgenes to specific genomic sites3,4. 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 using homology-directed repair5,6. Here we developed a CRISPR–Cas9 genome-targeting system that does not require viral vectors, allowing rapid and efficient insertion of large DNA sequences (greater than one kilobase) at specific sites in the genomes of primary human T cells, while preserving cell viability and function. This permits individual or multiplexed modification of endogenous genes. First, we applied this strategy to correct a pathogenic IL2RA mutation in cells from patients with monogenic autoimmune disease, and demonstrate improved signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR that redirected T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized tumour antigens and mounted productive anti-tumour cell responses in vitro and in vivo. Together, these studies provide preclinical evidence that non-viral genome targeting can enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.A non-viral strategy to introduce large DNA sequences into T cells enables the correction of a pathogenic mutation that causes autoimmunity, and the replacement of an endogenous T-cell receptor with an engineered receptor that can recognize cancer antigens.

[1]  F. Cosset,et al.  Lentiviral vector gene transfer into human T cells. , 2009, Methods in molecular biology.

[2]  Sruthi Mantri,et al.  CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells , 2016, Nature.

[3]  M. Toda,et al.  Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. , 1995, Journal of immunology.

[4]  Claudio Mussolino,et al.  Refining strategies to translate genome editing to the clinic , 2017, Nature Medicine.

[5]  S. Warming,et al.  Efficient mapping of transgene integration sites and local structural changes in Cre transgenic mice using targeted locus amplification , 2017, Nucleic acids research.

[6]  H. Dadi,et al.  Human immune disorder arising from mutation of the α chain of the interleukin-2 receptor , 1997 .

[7]  Bent K. Jakobsen,et al.  Single and Dual Amino Acid Substitutions in TCR CDRs Can Enhance Antigen-Specific T Cell Functions , 2008, The Journal of Immunology.

[8]  S. Rosenberg,et al.  High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[9]  Andreas Reinisch,et al.  Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6 , 2017, eLife.

[10]  Graham Dellaire,et al.  Marker-free coselection for CRISPR-driven genome editing in human cells , 2017, Nature Methods.

[11]  Jennifer A. Doudna,et al.  Generation of knock-in primary human T cells using Cas9 ribonucleoproteins , 2015, Proceedings of the National Academy of Sciences.

[12]  J. Murnane,et al.  Recombination events during integration of transfected DNA into normal human cells. , 1990, Nucleic acids research.

[13]  S. Henikoff,et al.  An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites , 2016, bioRxiv.

[14]  V. Hornung,et al.  Intracellular DNA recognition , 2010, Nature Reviews Immunology.

[15]  Bo Huang,et al.  A scalable strategy for high-throughput GFP tagging of endogenous human proteins , 2016, Proceedings of the National Academy of Sciences.

[16]  A. Scharenberg,et al.  Therapeutic Gene Editing Safety and Specificity. , 2017, Hematology/oncology clinics of North America.

[17]  Edward M. Callaway,et al.  In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration , 2016, Nature.

[18]  W. Lim,et al.  The Principles of Engineering Immune Cells to Treat Cancer , 2017, Cell.

[19]  David A. Scott,et al.  Double Nicking by RNA-Guided CRISPR Cas9 for Enhanced Genome Editing Specificity , 2013, Cell.

[20]  Mithat Gönen,et al.  Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection , 2017, Nature.

[21]  Howard Y. Chang,et al.  Discovery of stimulation-responsive immune enhancers with CRISPR activation , 2017, Nature.

[22]  Yutaka Inagaki,et al.  Easi-CRISPR: a robust method for one-step generation of mice carrying conditional and insertion alleles using long ssDNA donors and CRISPR ribonucleoproteins , 2017, Genome Biology.

[23]  A. Scharenberg,et al.  Homology-Directed Recombination for Enhanced Engineering of Chimeric Antigen Receptor T Cells , 2017, Molecular therapy. Methods & clinical development.

[24]  S. Rosenberg,et al.  Adoptive cell transfer as personalized immunotherapy for human cancer , 2015, Science.

[25]  Jonathan S. Weissman,et al.  Design and specificity of long ssDNA donors for CRISPR-based knock-in , 2017, bioRxiv.

[26]  Daesik Kim,et al.  Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins , 2014, Genome research.

[27]  M. Daly,et al.  Genetic and Epigenetic Fine-Mapping of Causal Autoimmune Disease Variants , 2014, Nature.

[28]  Michel Sadelain,et al.  Therapeutic T cell engineering , 2017, Nature.

[29]  G. Church,et al.  Cas9 as a versatile tool for engineering biology , 2013, Nature Methods.