Skip to main content

Advertisement

Log in

A multiplexed microfluidic continuous-flow electroporation system for efficient cell transfection

  • Research
  • Published:
Biomedical Microdevices Aims and scope Submit manuscript

Abstract

Cellular therapies have the potential to advance treatment for a broad array of diseases but rely on viruses for genetic reprogramming. The time and cost required to produce viruses has created a bottleneck that constricts development of and access to cellular therapies. Electroporation is a non-viral alternative for genetic reprogramming that bypasses these bottlenecks, but current electroporation technology suffers from low throughput, tedious optimization, and difficulty scaling to large-scale cell manufacturing. Here, we present an adaptable microfluidic electroporation platform with the capability for rapid, multiplexed optimization with 96-well plates. Once parameters are optimized using small volumes of cells, transfection can be seamlessly scaled to high-volume cell manufacturing without re-optimization. We demonstrate optimizing transfection of plasmid DNA to Jurkat cells, screening hundreds of different electrical waveforms of varying shapes at a speed of ~3 s per waveform using ~20 µL of cells per waveform. We selected an optimal set of transfection parameters using a low-volume flow cell. These parameters were then used in a separate high-volume flow cell where we obtained similar transfection performance by design. This demonstrates an alternative non-viral and economical transfection method for scaling to the volume required for producing a cell therapy without sacrificing performance. Importantly, this transfection method is disease-agnostic with broad applications beyond cell therapy.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author, HGC, upon reasonable request.

References

  • M. Bozza et al., A nonviral, nonintegrating DNA nanovector platform for the safe, rapid, and persistent manufacture of recombinant T cells. Sci. Adv. 7(16), 1–14 (2021)

    Article  Google Scholar 

  • J.P. Brody, P. Yager, R.E. Goldstein, R.H. Austin, Biotechnology at low Reynolds numbers. Biophys. J. 71(6), 3430–3441 (1996)

  • E. Buzhor et al., Cell-based therapy approaches: the hope for incurable diseases. Regen. Med. 9(5), 14–35 (2014)

    Article  Google Scholar 

  • B. Byhansonwade, Beacon adoptive cell: The current landscale (2023)

  • E. Capra, A. Gennari, C. Temps, viral-vector therapies at scale: Today’s challenges and future opportunities. (2022)

  • M. Cerrano et al., The advent of CAR T-cell therapy for lymphoproliferative neoplasms: integrating research into clinical practice. Front. Immunol. 11, 1–25 (2020)

  • S. Depil, P. Duchateau, S.A. Grupp, G. Mufti, L. Poirot, ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat. Rev. Drug Discov. (Nature Research) 19(3), 185–199 (2020)

  • S. Grupp et al., Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 368, 1509–1518 (2013)

  • T. Ingegnere et al., Human CAR NK cells: A new non-viral method allowing high efficient transfection and strong tumor cell killing. Front. Immunol. 10(APR), 1–10 (2019)

  • C.H. June, R.S. O’Connor, O.U. Kawalekar, S. Ghassemi, M.C. Milone, CAR T cell immunotherapy for human cancer. Science (American Association for the Advancement of Science) 359(6382), 1361–1365 (2018)

  • M. Kanduser, D. Miklavcic, M. Pavlin, Mechanisms involved in gene electrotransfer using high- and low-voltage pulses — An in vitro study. Bioelectrochemistry 74(2), 265–271 (2009)

    Article  Google Scholar 

  • T. Kotnik, G. Pucihar, M. Reberšek, D. Miklavčič, L.M. Mir, Role of pulse shape in cell membrane electropermeabilization. Biochim. Biophys. Acta Biomembr. 1614(2), 193–200 (2003)

  • T. Kotnik, L. Rems, M. Tarek, D. Miklavcic, Membrane electroporation and electropermeabilization: mechanisms and models. Annu. Rev. Biophys. 48, 63–91 (2019)

  • B.L. Levine, J. Miskin, K. Wonnacott, C. Keir, Global manufacturing of CAR T cell therapy. Mol. Ther. Methods Clin. Dev. 4(March), 92–101 (2017)

  • X. Liu et al., Novel T cells with improved in vivo anti-tumor activity generated by RNA electroporation. Protein Cell 8(7), 514–526 (2017)

    Article  Google Scholar 

  • C.A. Lissandrello, et al., High‑throughput continuous‑flow microfluidic electroporation of MRNA into primary human T cells for applications in cellular therapy manufacturing. Sci. Rep. 10, 1–16 (2020)

  • M.V. Maus, J.A. Fraietta, B.L. Levine, M. Kalos, Y. Zhao, C.H. June, Adoptive immunotherapy for cancer or viruses. Annu. Rev. Immunol. 32, 189–225 (2014)

  • N.M. Mount, S.J. Ward, P. Kefalas, J. Hyllner, N.M. Mount, Cell-based therapy technology classifications and translational challenges. Philos. Trans. B 370(20150017) (2015)

  • D. Porter, B. Levine, M. Kalos, A. Bagg, C.H. June, Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011)

  • S. Rafiq, C.S. Hackett, R.J. Brentjens, Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17(March), 147–167 (2020)

    Article  Google Scholar 

  • R.P.T. Somerville, L. Devillier, M.R. Parkhurst, S.A. Rosenberg, M.E. Dudley, Clinical scale rapid expansion of lymphocytes for adoptive cell transfer therapy in the WAVE ®bioreactor. J. Transl. Med. 10(1), 69 (2012)

    Article  Google Scholar 

  • M.P. Stewart, R. Langer, K.F. Jensen, Intracellular delivery by membrane disruption: Mechanisms, strategies, and concepts. Chem. Rev. 118(16), 7409–7531 (2018)

    Article  Google Scholar 

  • S.I. Sukharev, V.A. Klenchin, S.M. Serov, L.V. Chernomordik, A. Chizmadzhev Yu, Electroporation and electrophoretic DNA transfer into cells. The effect of DNA interaction with electropores. Biophys. J. 63(5), 1320–1327 (1992)

  • S. Sun, H. Hao, G. Yang, Y. Zhang, Y. Fu, Immunotherapy with CAR-modified T cells: toxicities and overcoming strategies. J. Immunol. Res. 2018, 1–10 (2018)

  • J. Teissie, M. Golzio, M.P. Rols, Mechanisms of cell membrane electropermeabilization: A minireview of our present (lack of ?) knowledge. Biochim. Biophys. Acta 1724, 270–280 (2005)

    Article  Google Scholar 

  • J. VanderBurgh, T. Corso, S. Levy, H. Craighead, Scalable continuous-flow electroporation platform enabling T cell transfection for cellular therapy manufacturing. Sci. Rep. 13 (2023)

  • J.C.M. Van Der Loo, J.F. Wright, Progress and challenges in viral vector manufacturing. Hum. Mol. Genet. 25, 42–52 (2016)

  • D.L. Wagner et al., Review: Sustainable clinical development of CAR-T cells – switching from viral transduction towards CRISPR-Cas gene editing. Front. Immunol. 13(June), 1–13 (2022)

  • J.C. Weaver, Y.A. Chizmadzhev, Theory of electroporation: A review. Bioelectrochemistry Bioenerg. 41, 135–160 (1996)

    Article  Google Scholar 

  • L. Wong, C. Brampton, D. Woo, E. Dreskin, Optimizing electroporation conditions for high-efficiency mRNA transfection of CD8+ T cells with the gene pulser xcell electroporation system mRNA using the gene pulser xcell electroporation system and gene pulser electroporation. (2020)

  • Y. Zhan et al., Low-frequency ac electroporation shows strong frequency dependence and yields comparable transfection results to dc electroporation. J. Control. Release 160(3), 570–576 (2012)

    Article  Google Scholar 

  • Z. Zhang, S. Qiu, X. Zhang, W. Chen, Optimized DNA electroporation for primary human T cell engineering. BMC Biotechnol. 18(1), 1–9 (2018)

    Article  Google Scholar 

Download references

Funding

This work was performed in part at the Cornell NanoScale Facility (CNF), a member of the National Nanotechnology Coordinated Infrastructure (NNCI), which is supported by the National Science Foundation (Grant NNCI-2025233). Research reported in this publication was supported by the National Institute of General Medical Sciences of the National Institutes of Health under Award Number R43GM148147. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: JAV, GTC, SLL, HGC. Methodology: JAV, GTC, SLL, HGC. Validation: JAV, GTC. Formal analysis: JAV. Investigation: JAV. Writing – original draft: JAV. Writing – review & editing: JAV, GTC, SLL, HGC. Visualization: JAV. Supervision: JAV, SLL, HGC. Project administration: JAV, SLL, HGC. Funding acquisition: JAV, SLL, HGC.

Corresponding author

Correspondence to Harold G. Craighead.

Ethics declarations

Competing interests

JAV, GTC, and HGC are listed as inventors on patent applications related to the technology presented and JAV, GTC, SLL, and HGC have a financial interest in CyteQuest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

VanderBurgh, J.A., Corso, G.T., Levy, S.L. et al. A multiplexed microfluidic continuous-flow electroporation system for efficient cell transfection. Biomed Microdevices 26, 10 (2024). https://doi.org/10.1007/s10544-023-00692-w

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s10544-023-00692-w

Keywords

Navigation