772
Views
8
CrossRef citations to date
0
Altmetric
Review

Physical transfection technologies for macrophages and dendritic cells in immunotherapy

ORCID Icon, , &
Pages 229-247 | Received 13 Aug 2020, Accepted 22 Sep 2020, Published online: 19 Oct 2020

References

  • Kim K-W, Kim S-H, Jang J-H, et al. Dendritic cells loaded with exogenous antigen by electroporation can enhance MHC class I–mediated antitumor immunity. Cancer Immunol Immunother. 2004;53:315–322.
  • Shaw CA, Starnbach MN. Stimulation of CD8+ T cells following diphtheria toxin-mediated antigen delivery into dendritic cells. Infect Immun. 2006;74:1001–1008.
  • Xu R-H, Remakus S, Ma X, et al. Direct presentation is sufficient for an efficient anti-viral CD8+ T cell response. PLoS Pathog. 2010;6:e1000768.
  • Foster S, Duvall CL, Crownover EF, et al. Intracellular delivery of a protein antigen with an endosomal-releasing polymer enhances CD8 T-cell production and prophylactic vaccine efficacy. Bioconjugate Chem. 2010;21:2205–2212.
  • Liu Z, Zhou C, Qin Y, et al. Coordinating antigen cytosolic delivery and danger signaling to program potent cross-priming by micelle-based nanovaccine. Cell Discov. 2017;3:1–14.
  • Klichinsky M, Ruella M, Shestova O, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38:947–953.
  • Kim CJ, Prevett T, Cormie J, et al. Dendritic cells infected with poxvin vitro iruses encoding MART-1/Melan A sensitize T lymphocytes in vitro. J Immunother. 1997;20:276–286.
  • Brossart P, Goldrath AW, Butz EA, et al. Virus-mediated delivery of antigenic epitopes into dendritic cells as a means to induce CTL. J Immunol. 1997;158:3270–3276.
  • Specht JM, Wang G, Do MT, et al. Dendritic cells retrovirally transduced with a model antigen gene are therapeutically effective against established pulmonary metastases. J Exp Med. 1997;186:1213–1221.
  • Dietz AB, Vuk-Pavlovic S. High efficiency adenovirus-mediated gene transfer to human dendritic cells. Blood. 1998;91:392–398.
  • Okada N, Saito T, Masunaga Y, et al. Efficient antigen gene transduction using arg-gly-asp fiber-mutant adenovirus vectors can potentiate antitumor vaccine efficacy and maturation of murine dendritic cells. Cancer Res. 2001;61:7913–7919.
  • Okada N, Iiyama S, Okada Y, et al. Immunological properties and vaccine efficacy of murine dendritic cells simultaneously expressing melanoma-associated antigen and interleukin-12. Cancer Gene Ther. 2005;12:72–83.
  • Morrissey MA, Williamson AP, Steinbach AM, et al. Chimeric antigen receptors that trigger phagocytosis. eLife. 2018;7:e36688.
  • Zhang L, Tian L, Dai X, et al. Induced pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions for liquid and solid tumors. bioRxiv 2020; 2020:03.28.011270.
  • Dullaers M, Breckpot K, Van Meirvenne S, et al. Side-by-side comparison of lentivirally transduced and mRNA-electroporated dendritic cells: implications for cancer immunotherapy protocols. Mol Ther. 2004;10:768–779.
  • Goyvaerts C, Breckpot K. The journey of in vivo virus engineered dendritic cells from bench to bedside: a bumpy road. Front Immunol. 2018;9:2052.
  • Breckpot K, Dullaers M, Bonehill A, et al. Lentivirally transduced dendritic cells as a tool for cancer immunotherapy. J Gene Med. 2003;5:654–667.
  • Leeuwen EBM, van Cloosen S, Senden-Gijsbers BLMG, et al. Transduction with a fiber-modified adenoviral vector is superior to non-viral nucleofection for expressing tumor-associated Ag mucin-1 in human DC. Cytotherapy. 2006;8:36–46.
  • Yin H, Kanasty RL, Eltoukhy AA, et al. Non-viral vectors for gene-based therapy. Nat Rev Genet. 2014;15:541–555.
  • Roth TL, Puig-Saus C, Yu R, et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature. 2018;559:405–409.
  • Lux CT, Scharenberg AM. Therapeutic gene editing safety and specificity. Hematol Oncol Clin North Am. 2017;31:787–795.
  • Stewart MP, Langer R, Jensen KF. Intracellular delivery by membrane disruption: mechanisms, strategies, and concepts. Chem Rev. 2018;118:7409–7531.
  • Hartman ZC, Appledorn DM, Amalfitano A. Adenovirus vector induced innate immune responses: impact upon efficacy and toxicity in gene therapy and vaccine applications. Virus Res. 2008;132:1–14.
  • Shirley JL, de Jong YP, Terhorst C, et al. Immune responses to viral gene therapy vectors. Mol Ther. 2020;28:709–722.
  • Muruve DA, Pétrilli V, Zaiss AK, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452:103–107.
  • Barlan AU, Griffin TM, Mcguire KA, et al. Adenovirus membrane penetration activates the NLRP3 inflammasome. J Virol. 2011;85:146–155.
  • Chen I-Y, Ichinohe T. Response of host inflammasomes to viral infection. Trends Microbiol. 2015;23:55–63.
  • Yazdani M, Jaafari MR, Verdi J, et al. Ex vivo-generated dendritic cell-based vaccines in melanoma: the role of nanoparticulate delivery systems. Immunotherapy. 2020;12:333–349.
  • Jensen K, Anderson JA, Glass EJ. Comparison of small interfering RNA (siRNA) delivery into bovine monocyte-derived macrophages by transfection and electroporation. Vet Immunol Immunopathol. 2014;158:224–232.
  • Verbeke R, Lentacker I, De Smedt SC, et al. Three decades of messenger RNA vaccine development. Nano Today. 2019;28:100766.
  • Van Tendeloo V, Snoeck H-W, Lardon F, et al. Nonviral transfection of distinct types of human dendritic cells: high-efficiency gene transfer by electroporation into hematopoietic progenitor- but not monocyte-derived dendritic cells. Gene Ther. 1998;5:700–707.
  • Strobel I, Berchtold S, Götze A, et al. Human dendritic cells transfected with either RNA or DNA encoding influenza matrix protein M1 differ in their ability to stimulate cytotoxic T lymphocytes. Gene Ther. 2000;7:2028–2035.
  • Zhong L, Granelli‐Piperno A, Choi Y, et al. Recombinant adenovirus is an efficient and non-perturbing genetic vector for human dendritic cells. Eur J Immunol. 1999;29:964–972.
  • Melhem NM, Gleason SM, Liu XD, et al. High-level antigen expression and sustained antigen presentation in dendritic cells nucleofected with wild-type viral mRNA but not DNA. Clin Vaccine Immunol. 2008;15:1337–1344.
  • Van Tendeloo VFI, Ponsaerts P, Lardon F, et al. Highly efficient gene delivery by mRNA electroporation in human hematopoietic cells: superiority to lipofection and passive pulsing of mRNA and to electroporation of plasmid cDNA for tumor antigen loading of dendritic cells. Blood. 2001;98:49–56.
  • Verbeke R, Lentacker I, Wayteck L, et al. Co-delivery of nucleoside-modified mRNA and TLR agonists for cancer immunotherapy: restoring the immunogenicity of immunosilent mRNA. J Control Release. 2017;266:287–300.
  • Firdessa-Fite R, Creusot RJ. Nanoparticles versus dendritic cells as vehicles to deliver mRNA encoding multiple epitopes for immunotherapy. Mol Ther Methods Clin Dev. 2020;16:50–62.
  • Xiang J, Xu L, Gong H, et al. Antigen-loaded upconversion nanoparticles for dendritic cell stimulation, tracking, and vaccination in dendritic cell-based immunotherapy. ACS Nano. 2015;9:6401–6411.
  • Dong X, Sun Z, Liang J, et al. A visible fluorescent nanovaccine based on functional genipin crosslinked ovalbumin protein nanoparticles. Nanomedicine. 2018;14:1087–1098.
  • Van Meirvenne S, Straetman L, Heirman C, et al. Efficient genetic modification of murine dendritic cells by electroporation with mRNA. Cancer Gene Ther. 2002;9:787–797.
  • Filion MC, Phillips NC. Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells. Biochim Biophys Acta - Biomembr. 1997;1329:345–356.
  • Lv H, Zhang S, Wang B, et al. Toxicity of cationic lipids and cationic polymers in gene delivery. J Control Release. 2006;114:100–109.
  • Jia J, Zhang Y, Xin Y, et al. Interactions between nanoparticles and dendritic cells: from the perspective of cancer immunotherapy. Front Oncol. 2018;8:404.
  • Zhong Z, Zhai Y, Liang S, et al. TRPM2 links oxidative stress to NLRP3 inflammasome activation. Nat Commun. 2013;4:1–11.
  • Meacham JM, Durvasula K, Degertekin FL, et al. Physical methods for intracellular delivery: practical aspects from laboratory use to industrial-scale processing. J Lab Autom. 2014;19:1–18.
  • Pelegrin P, Barroso-Gutierrez C, Surprenant A. P2X7 receptor differentially couples to distinct release pathways for IL-1β in mouse macrophage. J Immunol. 2008;180:7147–7157.
  • Chamberlain LM, Godek ML, Gonzalez‐Juarrero M, et al. Phenotypic non-equivalence of murine (monocyte-) macrophage cells in biomaterial and inflammatory models. J Biomed Mater Res A. 2009;88A:858–871.
  • Stacey KJ, Ross IL, Hume DA. Electroporation and DNA-dependent cell death in murine macrophages. Immunol Cell Biol. 1993;71:75–85.
  • Janeway CA, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216.
  • Hoebe K, Janssen E, Beutler B. The interface between innate and adaptive immunity. Nat Immunol. 2004;5:971–974.
  • Pozzi L-AM, Maciaszek JW, Rock KL. Both dendritic cells and macrophages can stimulate naive CD8 T cells in vivo to proliferate, develop effector function, and differentiate into memory cells. J Immunol. 2005;175:2071–2081.
  • Trombetta ES, Mellman I. Cell biology of antigen processing in vitro and in vivo. Annu Rev Immunol. 2005;23:975–1028.
  • Neefjes J, Jongsma MLM, Paul P, et al. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 2011;11:823–836.
  • Blum JS, Wearsch PA, Cresswell P. Pathways of antigen processing. Annu Rev Immunol. 2013;31:443–473.
  • Mellman I, Turley SJ, Steinman RM. Antigen processing for amateurs and professionals. Trends Cell Biol. 1998;8:231–237.
  • Cybulsky Myron I, Cheolho C, Robbins Clinton S. Macrophages and dendritic cells. Circ Res. 2016;118:637–652.
  • Di Pucchio T, Chatterjee B, Smed-Sörensen A, et al. Direct proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility complex class I. Nat Immunol. 2008;9:551–557.
  • Joffre OP, Segura E, Savina A, et al. Cross-presentation by dendritic cells. Nat Rev Immunol. 2012;12:557–569.
  • Gros M, Amigorena S. Regulation of antigen export to the cytosol during cross-presentation. Front Immunol. 2019;10:41.
  • Dhodapkar MV, Steinman RM, Krasovsky J, et al. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001;193:233–238.
  • Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045.
  • Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5:987–995.
  • Takeda K, Akira S. Toll-like receptors in innate immunity. Int Immunol. 2005;17:1–14.
  • Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801.
  • Diebold SS, Brencicova E. Nucleic acids and endosomal pattern recognition: how to tell friend from foe? Front Cell Infect Microbiol. 2013;3:37.
  • Hagar JA, Powell DA, Aachoui Y, et al. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science. 2013;341:1250–1253.
  • Saito T, Gale M. Differential recognition of double-stranded RNA by RIG-I–like receptors in antiviral immunity. J Exp Med. 2008;205:1523–1527.
  • Lage SL, Buzzo CL, Amaral EP, et al. Cytosolic flagellin-induced lysosomal pathway regulates inflammasome-dependent and -independent macrophage responses. PNAS. 2013;110:E3321–E3330.
  • Atianand MK, Fitzgerald KA. Molecular basis of DNA recognition in the immune system. J Immunol. 2013;190:1911–1918.
  • Liu D, Rhebergen AM, Eisenbarth SC. Licensing adaptive immunity by NOD-like receptors. Front Immunol. 2013;4:486.
  • Lamkanfi M, Dixit VM. The inflammasome turns 15. Nature. 2017;548:534–535.
  • Lamkanfi M. Emerging inflammasome effector mechanisms. Nat Rev Immunol. 2011;11:213–220.
  • Steinman RM, Cohn ZA. IDENTIFICATION OF A NOVEL CELL TYPE IN PERIPHERAL LYMPHOID ORGANS OF MICE I. MORPHOLOGY, QUANTITATION, TISSUE DISTRIBUTION. J Exp Med. 1973;137:1142–1162.
  • Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252.
  • Kaufmann SHE. Immunology’s foundation: the 100-year anniversary of the Nobel Prize to Paul Ehrlich and Elie Metchnikoff. Nat Immunol. 2008;9:705–712.
  • Kou PM, Babensee JE. Macrophage and dendritic cell phenotypic diversity in the context of biomaterials. J Biomed Mater Res A. 2011;96A:239–260.
  • Mills CD, Kincaid K, Alt JM, et al. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164:6166–6173.
  • Benoit M, Desnues B, Mege J-L. Macrophage polarization in bacterial infections. J Immunol. 2008;181:3733–3739.
  • Mills C. M1 and M2 macrophages: oracles of health and disease. CRI. 2012;32:463–488.
  • Schroder K, Hertzog PJ, Ravasi T, et al. Interferon-γ: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75:163–189.
  • Müller E, Christopoulos PF, Halder S, et al. Toll-like receptor ligands and interferon-γ synergize for induction of antitumor M1 macrophages. Front Immunol. 2017;8:1383.
  • Andreesen R, Hennemann B, Krause SW. Adoptive immunotherapy of cancer using monocyte-derived macrophages: rationale, current status, and perspectives. J Leukoc Biol. 1998;64:419–426.
  • Lee S, Kivimäe S, Dolor A, et al. Macrophage-based cell therapies: the long and winding road. J Control Release. 2016;240:527–540.
  • Ley K. M1 means kill; M2 means heal. J Immunol. 2017;199:2191–2193.
  • Delamarre L, Pack M, Chang H, et al. Differential lysosomal proteolysis in antigen-presenting cells determines antigen fate. Science. 2005;307:1630–1634.
  • Pauwels A-M, Trost M, Beyaert R, et al. Patterns, receptors, and signals: regulation of phagosome maturation. Trends Immunol. 2017;38:407–422.
  • Hume DA. Macrophages as APC and the dendritic cell myth. J Immunol. 2008;181:5829–5835.
  • Hashimoto D, Miller J, Merad M. Dendritic cell and macrophage heterogeneity in vivo. Immunity. 2011;35:323–335.
  • Peng M, Mo Y, Wang Y, et al. Neoantigen vaccine: an emerging tumor immunotherapy. Mol Cancer. 2019;18:128.
  • Filley AC, Dey M. Dendritic cell based vaccination strategy: an evolving paradigm. J Neurooncol. 2017;133:223–235.
  • Guo Y, Lei K, Tang L. Neoantigen vaccine delivery for personalized anticancer immunotherapy. Front Immunol. 2018;9:1499.
  • Steinman RM, Dhodapkar M. Active immunization against cancer with dendritic cells: the near future. Int J Cancer. 2001;94:459–473.
  • Zhu G, Zhang F, Ni Q, et al. Efficient nanovaccine delivery in cancer immunotherapy. ACS Nano. 2017;11:2387–2392.
  • Saxena M, Balan S, Roudko V, et al. Towards superior dendritic-cell vaccines for cancer therapy. Nat Biomed Eng. 2018;2:341–346.
  • Jonuleit H, Giesecke‐Tuettenberg A, Tüting T, et al. A comparison of two types of dendritic cell as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int J Cancer. 2001;93:243–251.
  • Wilgenhof S, Van Nuffel AMT, Benteyn D, et al. A phase IB study on intravenous synthetic mRNA electroporated dendritic cell immunotherapy in pretreated advanced melanoma patients. Ann Oncol. 2013;24:2686–2693.
  • Anguille S, Smits EL, Lion E, et al. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 2014;15:e257–e267.
  • Benteyn D, Heirman C, Bonehill A, et al. mRNA-based dendritic cell vaccines. Expert Rev Vaccines. 2015;14:161–176.
  • Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179:1109–1118.
  • Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005;5:296–306.
  • Lutz MB, Kukutsch N, Ogilvie ALJ, et al. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods. 1999;223:77–92.
  • Madaan A, Verma R, Singh AT, et al. A stepwise procedure for isolation of murine bone marrow and generation of dendritic cells. J Biol Methods. 2014;1:e1.
  • Turnis ME, Rooney CM. Enhancement of dendritic cells as vaccines for cancer. Immunotherapy. 2010;2:847–862.
  • Nair SK, Morse M, Boczkowski D, et al. Induction of tumor-specific cytotoxic T lymphocytes in cancer patients by autologous tumor RNA-transfected dendritic cells. Ann Surg. 2002;235:540–549.
  • Liu MA. A comparison of plasmid DNA and mRNA as vaccine technologies. Vaccines (Basel). 2019;7:37.
  • Lint SV, Heirman C, Thielemans K, et al. mRNA. Hum Vaccin Immunother. 2013;9:265–274.
  • Kamigaki T, Kaneko T, Naitoh K, et al. Immunotherapy of autologous tumor lysate-loaded dendritic cell vaccines by a closed-flow electroporation system for solid tumors. Anticancer Res. 2013;33:2971–2976.
  • Schreibelt G, Benitez-Ribas D, Schuurhuis D, et al. Commonly used prophylactic vaccines as an alternative for synthetically produced TLR ligands to mature monocyte-derived dendritic cells. Blood. 2010;116:564–574.
  • Castiello L, Sabatino M, Jin P, et al. Monocyte-derived DC maturation strategies and related pathways: a transcriptional view. Cancer Immunol Immunother. 2011;60:457–466.
  • Bonehill A, Tuyaerts S, Van Nuffel AM, et al. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Mol Ther. 2008;16:1170–1180.
  • Vries IJM, de Krooshoop DJEB, Scharenborg NM, et al. Effective migration of antigen-pulsed dendritic cells to lymph nodes in melanoma patients is determined by their maturation state. Cancer Res. 2003;63:12–17.
  • Hobo W, Maas F, Adisty N, et al. siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen–specific CD8+ T cells. Blood. 2010;116:4501–4511.
  • Van Lint S, Wilgenhof S, Heirman C, et al. Optimized dendritic cell-based immunotherapy for melanoma: the TriMix-formula. Cancer Immunol Immunother. 2014;63:959–967.
  • Harizaj A, De Clercq OQ, Descamps B, et al. Biocompatible lipid-coated persistent luminescent nanoparticles for in vivo imaging of dendritic cell migration. Part Part Syst Charact. 2019;36:1900371.
  • Nicolas T, Franck P, Arnaud M, et al. Adoptive immunotherapy for superficial bladder cancer with autologous macrophage activated killer cells. J Urol. 2002;168:2373–2376.
  • Pagès F, Lebel‐Binay S, Vieillefond A, et al. Local immunostimulation induced by intravesical administration of autologous interferon-gamma-activated macrophages in patients with superficial bladder cancer. Clin Exp Immunol. 2002;127:303–309.
  • Burger M, Thiounn N, Denzinger S, et al. The application of adjuvant autologous antravesical macrophage cell therapy vs. BCG in non-muscle invasive bladder cancer: a multicenter, randomized trial. J Transl Med. 2010;8:54.
  • Rems L. Chapter one - applicative use of electroporation models: from the molecular to the tissue level. Adv Biomembr Lipid Self-Assembly. 2017;26:1–50.
  • Kotnik T, Rems L, Tarek M, et al. Membrane electroporation and electropermeabilization: mechanisms and models. Annu Rev Biophys. 2019;48:63–91.
  • Saulis G, Venslauskas MS, Naktinis J. Kinetics of pore resealing in cell membranes after electroporation. J Electroanal Chem Interfacial Electrochem. 1991;321: 1–13.
  • Pucihar G, Kotnik T, Miklavčič D, et al. Kinetics of transmembrane transport of small molecules into electropermeabilized cells. Biophys J. 2008;95:2837–2848.
  • Sæbøe-Larssen S, Fossberg E, Gaudernack G. mRNA-based electrotransfection of human dendritic cells and induction of cytotoxic T lymphocyte responses against the telomerase catalytic subunit (hTERT). J Immunol Methods. 2002;259:191–203.
  • Landi A, Babiuk LA, Hurk S, et al. High transfection efficiency, gene expression, and viability of monocyte-derived human dendritic cells after nonviral gene transfer. J Leukoc Biol. 2007;82:849–860.
  • Lenz P, Bacot SM, Frazier-Jessen MR, et al. Nucleoporation of dendritic cells: efficient gene transfer by electroporation into human monocyte-derived dendritic cells. FEBS Lett. 2003;538:149–154.
  • Koski GK, Karikó K, Xu S, et al. Cutting edge: innate immune system discriminates between RNA containing bacterial versus eukaryotic structural features that prime for high-level IL-12 secretion by dendritic cells. J Immunol. 2004;172:3989–3993.
  • Karikó K, Buckstein M, Ni H, et al. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity. 2005;23:165–175.
  • Youn H, Chung J-K. Modified mRNA as an alternative to plasmid DNA (pDNA) for transcript replacement and vaccination therapy. Expert Opin Biol Ther. 2015;15:1337–1348.
  • Grünebach F, Müller MR, Nencioni A, et al. Delivery of tumor-derived RNA for the induction of cytotoxic T-lymphocytes. Gene Ther. 2003;10:367–374.
  • Van Meirvenne S, Dullaers M, Heirman C, et al. In vivo depletion of CD4+CD25+ regulatory T cells enhances the antigen-specific primary and memory CTL response elicited by mature mRNA-electroporated dendritic cells. Mol Ther. 2005;12:922–932.
  • Ponsaerts P, Van Tendeloo V, Cools N, et al. mRNA-electroporated mature dendritic cells retain transgene expression, phenotypical properties and stimulatory capacity after cryopreservation. Leukemia. 2002;16:1324–1330.
  • Tuyaerts S, Michiels A, Corthals J, et al. Induction of influenza matrix protein 1 and MelanA-specific T lymphocytes in vitro using mRNA-electroporated dendritic cells. Cancer Gene Ther. 2003;10:696–706.
  • Michiels A, Tuyaerts S, Bonehill A, et al. Electroporation of immature and mature dendritic cells: implications for dendritic cell-based vaccines. Gene Ther. 2005;12:772–782.
  • Javorovic M, Pohla H, Frankenberger B, et al. RNA transfer by electroporation into mature dendritic cells leading to reactivation of effector-memory cytotoxic T lymphocytes: a quantitative analysis. Mol Ther. 2005;12:734–743.
  • Schaft N, Dörrie J, Thumann P, et al. Generation of an optimized polyvalent monocyte-derived dendritic cell vaccine by transfecting defined RNAs after rather than before maturation. J Immunol. 2005;174:3087–3097.
  • Markovic SN, Dietz AB, Greiner CW, et al. Preparing clinical-grade myeloid dendritic cells by electroporation-mediated transfection of in vitro amplified tumor-derived mRNA and safety testing in stage IV malignant melanoma. J Transl Med. 2006;4:35.
  • Ö M, Eriksen J, Svane IM. Studies on mRNA electroporation of immature and mature dendritic cells: effects on their immunogenic potential. Mol Biotechnol. 2008;40:151–160.
  • Brabants E, Heyns K, Smet SD, et al. An accelerated, clinical-grade protocol to generate high yields of type 1-polarizing messenger RNA–loaded dendritic cells for cancer vaccination. Cytotherapy. 2018;20:1164–1181.
  • Chung DJ, Romano E, Pronschinske KB, et al. Langerhans-type and monocyte-derived human dendritic cells have different susceptibilities to mRNA electroporation with distinct effects on maturation and activation: implications for immunogenicity in dendritic cell-based immunotherapy. J Transl Med. 2013;11:166.
  • Driessche AV, Velde ALRV, de Nijs G, et al. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy. 2009;11:653–668.
  • Van Nuffel AM, Benteyn D, Wilgenhof S, et al. Dendritic cells loaded with mRNA encoding full-length tumor antigens prime CD4+ and CD8+ T cells in melanoma patients. Mol Ther. 2012;20:1063–1074.
  • Hobo W, Novobrantseva TI, Fredrix H, et al. Improving dendritic cell vaccine immunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles combined with antigen mRNA electroporation. Cancer Immunol Immunother. 2013;62:285–297.
  • Prechtel AT, Turza NM, Theodoridis AA, et al. Small interfering RNA (siRNA) delivery into monocyte-derived dendritic cells by electroporation. J Immunol Methods. 2006;311:139–152.
  • Jantsch J, Turza N, Volke M, et al. Small interfering RNA (siRNA) delivery into murine bone marrow-derived dendritic cells by electroporation. J Immunol Methods. 2008;337:71–77.
  • Schuurhuis DH, Verdijk P, Schreibelt G, et al. In situ expression of tumor antigens by messenger RNA–electroporated dendritic cells in lymph nodes of melanoma patients. Cancer Res. 2009;69:2927–2934.
  • Morse MA, Mosca PJ, Clay TM, et al. Dendritic cell maturation in active immunotherapy strategies. Expert Opin Biol Ther. 2002;2:35–43.
  • Zhang M, Ma Z, Selliah N, et al. The impact of nucleofection® on the activation state of primary human CD4 T cells. J Immunol Methods. 2014;408:123–131.
  • DiTommaso T, Cole JM, Cassereau L, et al. Cell engineering with microfluidic squeezing preserves functionality of primary immune cells in vivo. PNAS. 2018;115:E10907–E10914.
  • Franz KM, Kagan JC. Innate immune receptors as competitive determinants of cell fate. Mol Cell. 2017;66:750–760.
  • Man SM, Karki R, Kanneganti T-D. DNA-sensing inflammasomes: regulation of bacterial host defense and the gut microbiota. Pathog Dis. 2016;74:ftw028.
  • Fernandes-Alnemri T, Yu J-W, Datta P, et al. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009;458:509–513.
  • Judge AD, Sood V, Shaw JR, et al. Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol. 2005;23:457–462.
  • Hornung V, Guenthner-Biller M, Bourquin C, et al. Sequence-specific potent induction of IFN-α by short interfering RNA in plasmacytoid dendritic cells through TLR7. Nat Med. 2005;11:263–270.
  • Wiese M, Castiglione K, Hensel M, et al. Small interfering RNA (siRNA) delivery into murine bone marrow-derived macrophages by electroporation. J Immunol Methods. 2010;353:102–110.
  • Lee BL, Mirrashidi KM, Stowe IB, et al. ASC- and caspase-8-dependent apoptotic pathway diverges from the NLRC4 inflammasome in macrophages. Sci Rep. 2018;8:37788.
  • Zhou Y, Yang K, Cui J, et al. Controlled permeation of cell membrane by single bubble acoustic cavitation. J Control Release. 2012;157:103–111.
  • Lentacker I, De Cock I, Deckers R, et al. Understanding ultrasound induced sonoporation: definitions and underlying mechanisms. Adv Drug Deliv Rev. 2014;72:49–64.
  • Suzuki R, Oda Y, Utoguchi N, et al. A novel strategy utilizing ultrasound for antigen delivery in dendritic cell-based cancer immunotherapy. J Control Release. 2009;133:198–205.
  • De Temmerman M-L, Dewitte H, Vandenbroucke RE, et al. mRNA-Lipoplex loaded microbubble contrast agents for ultrasound-assisted transfection of dendritic cells. Biomaterials. 2011;32:9128–9135.
  • Dewitte H, Van Lint S, Heirman C, et al. The potential of antigen and TriMix sonoporation using mRNA-loaded microbubbles for ultrasound-triggered cancer immunotherapy. J Control Release. 2014;194:28–36.
  • Lemmon JCM, McFarland RJ, Rybicka JM, et al. In vitro and in vivo transfection of primary phagocytes via microbubble-mediated intraphagosomal sonoporation. J Immunol Methods. 2011;371:152–158.
  • Kim W, Ng JK, Kunitake ME, et al. Interfacing silicon nanowires with mammalian cells. J Am Chem Soc. 2007;129:7228–7229.
  • Hällström W, Mårtensson T, Prinz C, et al. Gallium phosphide nanowires as a substrate for cultured neurons. Nano Lett. 2007;7:2960–2965.
  • Shalek AK, Robinson JT, Karp ES, et al. Vertical silicon nanowires as a universal platform for delivering biomolecules into living cells. PNAS. 2010;107:1870–1875.
  • Shalek AK, Gaublomme JT, Wang L, et al. Nanowire-mediated delivery enables functional interrogation of primary immune cells: application to the analysis of chronic lymphocytic leukemia. Nano Lett. 2012;12:6498–6504.
  • Xie X, Xu AM, Angle MR, et al. Mechanical model of vertical nanowire cell penetration. Nano Lett. 2013;13:6002–6008.
  • Xie X, Aalipour A, Gupta SV, et al. Determining the time window for dynamic nanowire cell penetration processes. ACS Nano. 2015;9:11667–11677.
  • Nair BG, Hagiwara K, Ueda M, et al. High density of aligned nanowire treated with polydopamine for efficient gene silencing by siRNA according to cell membrane perturbation. ACS Appl Mater Interfaces. 2016;8:18693–18700.
  • Mumm F, Beckwith KM, Bonde S, et al. A transparent nanowire-based cell impalement device suitable for detailed cell–nanowire interaction studies. Small. 2013;9:263–272.
  • Berthing T, Bonde S, Rostgaard KR, et al. Cell membrane conformation at vertical nanowire array interface revealed by fluorescence imaging. Nanotechnology. 2012;23:415102.
  • Robinson JT, Jorgolli M, Shalek AK, et al. Vertical nanowire electrode arrays as a scalable platform for intracellular interfacing to neuronal circuits. Nat Nanotechnol. 2012;7:180–184.
  • Persson H, Købler C, Mølhave K, et al. Fibroblasts cultured on nanowires exhibit low motility, impaired cell division, and DNA damage. Small. 2013;9:4006–4016.
  • Sharei A, Zoldan J, Adamo A, et al. A vector-free microfluidic platform for intracellular delivery. PNAS. 2013;110:2082–2087.
  • Sharei A, Trifonova R, Jhunjhunwala S, et al. Ex vivo cytosolic delivery of functional macromolecules to immune cells. PLoS One. 2015;10:e0118803.
  • Sharei A, Poceviciute R, Jackson EL, et al. Plasma membrane recovery kinetics of a microfluidic intracellular delivery platform. Int Bio (Cam). 2014;6:470–475.
  • Griesbeck M, Ziegler S, Laffont S, et al. Sex differences in plasmacytoid dendritic cell levels of IRF5 drive higher IFN-α production in women. J Immunol. 2015;195:5327–5336.
  • Deng Y, Kizer M, Rada M, et al. Intracellular delivery of nanomaterials via an inertial microfluidic cell hydroporator. Nano Lett. 2018;18:2705–2710.
  • Kizer ME, Deng Y, Kang G, et al. Hydroporator: a hydrodynamic cell membrane perforator for high-throughput vector-free nanomaterial intracellular delivery and DNA origami biostability evaluation. Lab Chip. 2019;19:1747–1754.
  • Jarrell JA, Twite AA, Lau KHWJ, et al. Intracellular delivery of mRNA to human primary T cells with microfluidic vortex shedding. Sci Rep. 2019;9:3214.
  • Xiong R, Raemdonck K, Peynshaert K, et al. Comparison of gold nanoparticle mediated photoporation: vapor nanobubbles outperform direct heating for delivering macromolecules in live cells. ACS Nano. 2014;8:6288–6296.
  • Xiong R, Drullion C, Verstraelen P, et al. Fast spatial-selective delivery into live cells. J Control Release. 2017;266:198–204.
  • Liu J, Li C, Brans T, et al. Surface functionalization with polyethylene glycol and polyethyleneimine improves the performance of graphene-based materials for safe and efficient intracellular delivery by laser-induced photoporation. Int J Mol Sci. 2020;21:1540.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.