Comparing intracellular delivery methods of electroporation versus cell squeezing (a microfluidic membrane deformation technique), DiTommaso et al. found that while the methods were similar in their ability to produce viable, edited T cells, electroporation disrupted gene expression profiles, affected a variety of cellular pathways, drove PD-1 upregulation, and induced non-specific cytokine bursts that ultimately dampened the long-term effector functions of the T cells and limited their antitumor efficacy in vivo. Squeezed T cells maintained a phenotypic profile similar to controls, and showed antitumor efficacy in mice.

The translational potential of cell-based therapies is often limited by complications related to effectively engineering and manufacturing functional cells. While the use of electroporation is widespread, the impact of electroporation on cell state and function has yet to be fully characterized. Here, we use a genome-wide approach to study optimized electroporation treatment and identify striking disruptions in the expression profiles of key functional transcripts of human T cells. These genetic disruptions result in concomitant perturbation of cytokine secretion including a 648-fold increase in IL-2 secretion (P < 0.01) and a 30-fold increase in IFN-gamma secretion (P < 0.05). Ultimately, the effects at the transcript and protein level resulted in functional deficiencies in vivo, with electroporated T cells failing to demonstrate sustained antigen-specific effector responses when subjected to immunological challenge. In contrast, cells subjected to a mechanical membrane disruption-based delivery mechanism, cell squeezing, had minimal aberrant transcriptional responses [0% of filtered genes misregulated, false discovery rate (FDR) q < 0.1] relative to electroporation (17% of genes misregulated, FDR q < 0.1) and showed undiminished effector responses, homing capabilities, and therapeutic potential in vivo. In a direct comparison of functionality, T cells edited for PD-1 via electroporation failed to distinguish from untreated controls in a therapeutic tumor model, while T cells edited with similar efficiency via cell squeezing demonstrated the expected tumor-killing advantage. This work demonstrates that the delivery mechanism used to insert biomolecules affects functionality and warrants further study.

Author Info: (1) SQZ Biotechnologies, Watertown, MA 02472; tia.ditommaso@sqzbiotech.com. (2) SQZ Biotechnologies, Watertown, MA 02472. (3) SQZ Biotechnologies, Watertown, MA 02472. (4) SQZ Biot

Author Info: (1) SQZ Biotechnologies, Watertown, MA 02472; tia.ditommaso@sqzbiotech.com. (2) SQZ Biotechnologies, Watertown, MA 02472. (3) SQZ Biotechnologies, Watertown, MA 02472. (4) SQZ Biotechnologies, Watertown, MA 02472. (5) SQZ Biotechnologies, Watertown, MA 02472. (6) SQZ Biotechnologies, Watertown, MA 02472. (7) SQZ Biotechnologies, Watertown, MA 02472. (8) SQZ Biotechnologies, Watertown, MA 02472. (9) SQZ Biotechnologies, Watertown, MA 02472. (10) SQZ Biotechnologies, Watertown, MA 02472. (11) Oncology Discovery Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Zurich, 8952 Schlieren, Switzerland. (12) Oncology Discovery Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Zurich, 8952 Schlieren, Switzerland. (13) Oncology Discovery Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Zurich, 8952 Schlieren, Switzerland. (14) Oncology Discovery Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Zurich, 8952 Schlieren, Switzerland. (15) Oncology Discovery Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Zurich, 8952 Schlieren, Switzerland. (16) SQZ Biotechnologies, Watertown, MA 02472. (17) SQZ Biotechnologies, Watertown, MA 02472.