Clinical Applications and Immunological Aspects of Electroporation-Based Therapies
Abstract
:1. Introduction
2. Plasma Membrane Electroporation
3. Clinical Aspects of EP-Related Therapies
3.1. Bleomycin
3.2. Electrochemotherapy
4. Immunological Aspects of Reversible Electroporation and Electrochemotherapy
5. Gene Electrotransfer (GET) and Electrogenechemotherapy (EGCT) Alone or in Combination with Other Immunotherapies
6. Variability of Responses in EP-Related Therapies
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Miklavcic, D.; Mir, L.M.; Thomas Vernier, P. Electroporation-based technologies and treatments. J. Membr. Biol. 2010, 236, 1–2. [Google Scholar] [CrossRef] [Green Version]
- Neumann, E.; Rosenheck, K. Permeability changes induced by electric impulses in vesicular membranes. J. Membr. Biol. 1972, 10, 279–290. [Google Scholar] [CrossRef] [Green Version]
- Neumann, E.; Schaefer-Ridder, M.; Wang, Y.; Hofschneider, P.H. Gene transfer into mouse lyoma cells by electroporation in high electric fields. EMBO J. 1982, 1, 841–845. [Google Scholar] [CrossRef]
- Geboers, B.; Scheffer, H.J.; Graybill, P.M.; Ruarus, A.H.; Nieuwenhuizen, S.; Puijk, R.S.; van den Tol, P.M.; Davalos, R.V.; Rubinsky, B.; de Gruijl, T.D.; et al. High-Voltage Electrical Pulses in Oncology: Irreversible Electroporation, Electrochemotherapy, Gene Electrotransfer, Electrofusion, and Electroimmunotherapy. Radiology 2020, 295, 254–272. [Google Scholar] [CrossRef] [PubMed]
- Tsong, T.Y. Electroporation of cell membranes. Biophys. J. 1991, 60, 297–306. [Google Scholar] [CrossRef] [Green Version]
- Rols, M.-P. Parameters Affecting Cell Viability Following Electroporation In Vitro; Springer: Cham, Switzerland, 2016; pp. 1–17. [Google Scholar]
- Tu, Q.; Yin, J.; Fu, J.; Herrmann, J.; Li, Y.; Yin, Y.; Stewart, A.F.; Muller, R.; Zhang, Y. Room temperature electrocompetent bacterial cells improve DNA transformation and recombineering efficiency. Sci. Rep. 2016, 6, 24648. [Google Scholar] [CrossRef] [PubMed]
- Dower, W.J.; Miller, J.F.; Ragsdale, C.W. High efficiency transformation of E. coli by high voltage electroporation. Nucleic Acids Res. 1988, 16, 6127–6145. [Google Scholar] [CrossRef] [Green Version]
- Neumann, E.; Sowers, A.E.; Jordan, C.A. Electroporation and Electrofusion in Cell Biology; Springer US: New York, NY, USA, 1989. [Google Scholar]
- Hu, N.; Yang, J.; Joo, S.W.; Banerjee, A.N.; Qian, S. Cell electrofusion in microfluidic devices: A review. Sens. Actuators B Chem. 2013, 178, 63–85. [Google Scholar] [CrossRef]
- Medi, B.M.; Layek, B.; Singh, J. Electroporation for Dermal and Transdermal Drug Delivery; Springer: Berlin/Heidelberg, Germany, 2017; pp. 105–122. [Google Scholar]
- Yarmush, M.L.; Golberg, A.; Sersa, G.; Kotnik, T.; Miklavcic, D. Electroporation-based technologies for medicine: Principles, applications, and challenges. Annu. Rev. Biomed. Eng. 2014, 16, 295–320. [Google Scholar] [CrossRef] [Green Version]
- Lu, Y.; Xue, J.; Deng, T.; Zhou, X.; Yu, K.; Deng, L.; Huang, M.; Yi, X.; Liang, M.; Wang, Y.; et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat. Med. 2020, 26, 732–740. [Google Scholar] [CrossRef]
- Chicaybam, L.; Abdo, L.; Viegas, M.; Marques, L.V.C.; de Sousa, P.; Batista-Silva, L.R.; Alves-Monteiro, V.; Bonecker, S.; Monte-Mor, B.; Bonamino, M.H. Transposon-mediated generation of CAR-T cells shows efficient anti B-cell leukemia response after ex vivo expansion. Gene Ther. 2020, 27, 85–95. [Google Scholar] [CrossRef] [PubMed]
- Smith, T.R.F.; Patel, A.; Ramos, S.; Elwood, D.; Zhu, X.; Yan, J.; Gary, E.N.; Walker, S.N.; Schultheis, K.; Purwar, M.; et al. Immunogenicity of a DNA vaccine candidate for COVID-19. Nat. Commun. 2020, 11, 2601. [Google Scholar] [CrossRef] [PubMed]
- Lambricht, L.; Lopes, A.; Kos, S.; Sersa, G.; Preat, V.; Vandermeulen, G. Clinical potential of electroporation for gene therapy and DNA vaccine delivery. Expert Opin. Drug Deliv. 2016, 13, 295–310. [Google Scholar] [CrossRef] [PubMed]
- Chiarella, P.; Michele, V.; Signori, E. DNA Vaccination by Electrogene Transfer. In Non-Viral Gene Therapy; Yuan, X., Ed.; IntechOpen: London, UK, 2011; pp. 169–198. ISBN 978-953-51-4408-3. [Google Scholar]
- Basu, G.; Downey, H.; Guo, S.; Israel, A.; Asmar, A.; Hargrave, B.; Heller, R. Prevention of distal flap necrosis in a rat random skin flap model by gene electro transfer delivering VEGF(165) plasmid. J. Gene Med. 2014, 16, 55–65. [Google Scholar] [CrossRef] [PubMed]
- Touchard, E.; Berdugo, M.; Bigey, P.; El Sanharawi, M.; Savoldelli, M.; Naud, M.C.; Jeanny, J.C.; Behar-Cohen, F. Suprachoroidal electrotransfer: A nonviral gene delivery method to transfect the choroid and the retina without detaching the retina. Mol. Ther. 2012, 20, 1559–1570. [Google Scholar] [CrossRef] [Green Version]
- Ferraro, B.; Cruz, Y.L.; Coppola, D.; Heller, R. Intradermal delivery of plasmid VEGF(165) by electroporation promotes wound healing. Mol. Ther. 2009, 17, 651–657. [Google Scholar] [CrossRef] [PubMed]
- Dezawa, M.; Takano, M.; Negishi, H.; Mo, X.; Oshitari, T.; Sawada, H. Gene transfer into retinal ganglion cells by in vivo electroporation: A new approach. Micron. Micron. 2002, 33, 1–6. [Google Scholar] [CrossRef]
- Mittal, L.; Camarillo, I.G.; Varadarajan, G.S.; Srinivasan, H.; Aryal, U.K.; Sundararajan, R. High-throughput, Label-Free Quantitative Proteomic Studies of the Anticancer Effects of Electrical Pulses with Turmeric Silver Nanoparticles: An in vitro Model Study. Sci Rep. 2020, 10, 7258. [Google Scholar] [CrossRef]
- Pasquet, L.; Bellard, E.; Chabot, S.; Markelc, B.; Rols, M.P.; Teissie, J.; Golzio, M. Pre-clinical investigation of the synergy effect of interleukin-12 gene-electro-transfer during partially irreversible electropermeabilization against melanoma. J. Immunother. Cancer 2019, 7, 161. [Google Scholar] [CrossRef] [Green Version]
- Marty, M.S.G.; Garbay, J.R.G.J.; Collins, C.G.S.M.; Billard, V.G.P.F.; Larkin, J.O.M.D.; Pavlovic, I.P.-K.S.M.; Cemazar, M.M.N.; Soden, D.M.R.Z.; Robert, C.O.S.G.C.; Mir, L.M. Electrochemotherapy-An easy, highly effective and safe treatment of cutaneous and subcutaneous metastases: Results of ESOPE (European Standard Operating Procedures of Electrochemotherapy) study. EJC Suppl. 2006, 4, 3. [Google Scholar] [CrossRef]
- Buckow, R.; Ng, S.; Toepfl, S. Pulsed Electric Field Processing of Orange Juice: A Review on Microbial, Enzymatic, Nutritional, and Sensory Quality and Stability. Compr. Rev. Food Sci. Food Saf. 2013, 12, 455–467. [Google Scholar] [CrossRef]
- Maged, E.A.M.; Ayman, H.A.E. Pulsed Electric Fields for Food Processing Technology; Intech Open Access Publisher: London, UK, 2012. [Google Scholar]
- Rieder, A.; Schwartz, T.; Schon-Holz, K.; Marten, S.M.; Suss, J.; Gusbeth, C.; Kohnen, W.; Swoboda, W.; Obst, U.; Frey, W. Molecular monitoring of inactivation efficiencies of bacteria during pulsed electric field treatment of clinical wastewater. J. Appl. Microbiol. 2008, 105, 2035–2045. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Wang, T.; Yu, C.; Xie, X. Locally enhanced electric field treatment (LEEFT) for water disinfection. Front. Environ. Sci. Eng. Front. Environ. Sci. Eng. 2020, 14, 78. [Google Scholar] [CrossRef]
- Kotnik, T.; Rems, L.; Tarek, M.; Miklavcic, D. Membrane Electroporation and Electropermeabilization: Mechanisms and Models. Annu. Rev. Biophys. 2019, 48, 63–91. [Google Scholar] [CrossRef]
- Cemazar, M.; Sersa, G.; Frey, W.; Miklavcic, D.; Teissie, J. Recommendations and requirements for reporting on applications of electric pulse delivery for electroporation of biological samples. Bioelectrochemistry 2018, 122, 69–76. [Google Scholar] [CrossRef]
- Spugnini, E.P.; Baldi, A. Electrochemotherapy in Veterinary Oncology: State-of-the-Art and Perspectives. Vet. Clin. North. Am. Small Anim. Pract. 2019, 49, 967–979. [Google Scholar] [CrossRef]
- Lee, E.W.; Thai, S.; Kee, S.T. Irreversible electroporation: A novel image-guided cancer therapy. Gut Liver 2010, 4, S99–S104. [Google Scholar] [CrossRef]
- Gissel, H.; Lee, R.C.; Gehl, J. Electroporation and Cellular Physiology; Springer: New York, NY, USA, 2011; pp. 9–17. [Google Scholar]
- Davalos, R.V.; Mir, I.L.; Rubinsky, B. Tissue ablation with irreversible electroporation. Ann. Biomed. Eng. 2005, 33, 223–231. [Google Scholar] [CrossRef] [PubMed]
- Beebe, S.J.; Sain, N.M.; Ren, W. Induction of Cell Death Mechanisms and Apoptosis by Nanosecond Pulsed Electric Fields (nsPEFs). Cells 2013, 2, 136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beebe, S.J.; White, J.; Blackmore, P.F.; Deng, Y.; Somers, K.; Schoenbach, K.H. Diverse effects of nanosecond pulsed electric fields on cells and tissues. DNA Cell Biol. 2003, 22, 785–796. [Google Scholar] [CrossRef] [PubMed]
- Cutrera, J.; King, G.; Jones, P.; Kicenuik, K.; Gumpel, E.; Xia, X.; Li, S. Safe and effective treatment of spontaneous neoplasms with interleukin 12 electro-chemo-gene therapy. J. Cell Mol. Med. 2015, 19, 664–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trainito, C. Study of Cell Membrane Permeabilization Induced by Pulsed Electric Field-Electrical Modeling and Characterization on Biochip; Univeristẻ Paris-Saclay: Paris, France, 2015. [Google Scholar]
- Alberts, B.J.A.; Lewis, J.; Morgan, D.; Raff, M.; Roberts, K.; Walter, P. Membrane transport of small molecules and the electrical properties of membranes. In Molecular Biology of the Cell; Garland Science: New York, NY, USA, 2015; pp. 597–640. [Google Scholar]
- Rems, L. Lipid Pores: Molecular and Continuum Models; Springer: Berlin/Heidelberg, Germany, 2016; pp. 1–21. [Google Scholar]
- Alberts, B.J.A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. The Universal Features of Cells on Earth. In Molecular Biology of the Cell; Garland Science: New York, NY, USA, 2015; pp. 2–10. [Google Scholar]
- Gehl, J.; Sersa, G.; Matthiessen, L.W.; Muir, T.; Soden, D.; Occhini, A.; Quaglino, P.; Curatolo, P.; Campana, L.G.; Kunte, C.; et al. Updated standard operating procedures for electrochemotherapy of cutaneous tumours and skin metastases. Acta Oncol. 2018, 57, 874–882. [Google Scholar] [CrossRef] [PubMed]
- Levine, Z.A.; Vernier, P.T. Life cycle of an electropore: Field-dependent and field-independent steps in pore creation and annihilation. J. Membr. Biol. 2010, 236, 27–36. [Google Scholar] [CrossRef]
- Hofmann, G.A. Instrumentation and electrodes for in vivo electroporation. Methods Mol. Med. 2000, 37, 37–61. [Google Scholar]
- Ruzgys, P.; Novickij, V.; Novickij, J.; Satkauskas, S. Nanosecond range electric pulse application as a non-viral gene delivery method: Proof of concept. Sci. Rep. 2018, 8, 15502. [Google Scholar] [CrossRef]
- Corovic, S.; Lackovic, I.; Sustaric, P.; Sustar, T.; Rodic, T.; Miklavcic, D. Modeling of electric field distribution in tissues during electroporation. Biomed. Eng. Online 2013, 12, 16. [Google Scholar] [CrossRef] [Green Version]
- Bisceglia, B.; Scaglione, A.; Tallarino, N.F.; Terlizzi, F.D. Electroporation of bone cancer: Numerical evaluation of the electric field in treated tissues. COMPEL Int. J. Comput. Math. Electr. Electron. Eng. 2013, 32, 1912–1928. [Google Scholar] [CrossRef]
- Teissie, J.; Golzio, M.; Rols, M.P. Mechanisms of cell membrane electropermeabilization: A minireview of our present (lack of ?) knowledge. Biochim. Biophys. Acta 2005, 1724, 270–280. [Google Scholar] [CrossRef]
- Cemazar, M.; Kotnik, T.; Sersa, G.; Miklavcic, D. Electroporation for electrochemotherapy and gene therapy. Electromagn. Fields Biol. Med. 2015, 24, 395–413. [Google Scholar]
- Jourabchi, N.; Beroukhim, K.; Tafti, B.A.; Kee, S.T.; Lee, E.W. Irreversible electroporation (NanoKnife) in cancer treatment. GII Gastrointest. Interv. 2014, 3, 8–18. [Google Scholar] [CrossRef] [Green Version]
- Miklavcic, D.; Kotnik, T. Electromagnetic Fields in Biology and Medicine; Markov, M.S., Ed.; CRC Press: Boca Raton, FL, USA, 2015. [Google Scholar]
- Mir, L.M.; Tounekti, O.; Orlowski, S. Bleomycin: Revival of an old drug. Gen. Pharmacol. Vasc. Syst. Gen. Pharmacol. Vasc. Syst. 1996, 27, 745–748. [Google Scholar] [CrossRef]
- Rangel, M.M.M.; Luz, J.C.S.; Oliveira, K.D.; Ojeda, J.; Freytag, J.O.; Suzuki, D.O. Electrochemotherapy in the treatment of neoplasms in dogs and cats. Austral. J. Vet. Sci. Austral. J. Vet. Sci. 2019, 51, 45–51. [Google Scholar] [CrossRef] [Green Version]
- Tozon, N.; Lampreht Tratar, U.; Znidar, K.; Sersa, G.; Teissie, J.; Cemazar, M. Operating Procedures of the Electrochemotherapy for Treatment of Tumor in Dogs and Cats. J. Vis. Exp. 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Landstrom, F.; Ivarsson, M.; Von Sydow, A.K.; Magnuson, A.; Von Beckerath, M.; Moller, C. Electrochemotherapy-Evidence for Cell-type Selectivity In Vitro. Anticancer. Res. 2015, 35, 5813–5820. [Google Scholar] [PubMed]
- Miklavcic, D.; Sersa, G.; Brecelj, E.; Gehl, J.; Soden, D.; Bianchi, G.; Ruggieri, P.; Rossi, C.R.; Campana, L.G.; Jarm, T. Electrochemotherapy: Technological advancements for efficient electroporation-based treatment of internal tumors. Med. Biol. Eng. Comput. 2012, 50, 1213–1225. [Google Scholar] [CrossRef] [Green Version]
- Jarm, T.; Cemazar, M.; Miklavcic, D.; Sersa, G. Antivascular effects of electrochemotherapy: Implications in treatment of bleeding metastases. Expert Rev. Anticancer Ther. 2010, 10, 729–746. [Google Scholar] [CrossRef] [PubMed]
- Mir, L.M. Bases and rationale of the electrochemotherapy. Eur. J. Cancer Suppl. 2006, 4, 38–44. [Google Scholar] [CrossRef]
- Tounekti, O.; Kenani, A.; Foray, N.; Orlowski, S.; Mir, L.M. The ratio of single- to double-strand DNA breaks and their absolute values determine cell death pathway. Br. J. Cancer 2001, 84, 1272–1279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hecht, S.M. Bleomycin: New perspectives on the mechanism of action. J. Nat. Prod. 2000, 63, 158–168. [Google Scholar] [CrossRef]
- Mekid, H.; Tounekti, O.; Spatz, A.; Cemazar, M.; El Kebir, F.Z.; Mir, L.M. In vivo evolution of tumour cells after the generation of double-strand DNA breaks. Br. J. Cancer 2003, 88, 1763–1771. [Google Scholar] [CrossRef] [Green Version]
- Tounekti, O.; Pron, G.; Belehradek, J.; Mir, L.M. Bleomycin, an Apoptosis-mimetic Drug That Induces Two Types of Cell Death Depending on the Number of Molecules Internalized. Cancer Res. 1993, 53, 5462. [Google Scholar]
- Calvet, C.Y.; Mir, L.M. The promising alliance of anti-cancer electrochemotherapy with immunotherapy. Cancer Metastasis Rev. 2016, 35, 165–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mir, L.M.; Gehl, J.; Sersa, G.; Collins, C.G.; Garbay, J.R.; Billard, V.; Geertsen, P.F.; Rudolf, Z.; O’Sullivan, G.C.; Marty, M. Standard operating procedures of the electrochemotherapy: Instructions for the use of bleomycin or cisplatin administered either systemically or locally and electric pulses delivered by the CliniporatorTM by means of invasive or non-invasive electrodes. Eur. J. Cancer Suppl. 2006, 4, 14–25. [Google Scholar] [CrossRef]
- Mir, L.M.; Orlowski, S.; Belehradek, J.; Paoletti, C. Electrochemotherapy potentiation of antitumour effect of bleomycin by local electric pulses. EJCCO Eur. J. Cancer Clin. Oncol. 1991, 27, 68–72. [Google Scholar] [CrossRef]
- Mir, L.M.; Belehradek, M.; Domenge, C.; Orlowski, S.; Poddevin, B.; Belehradek, J., Jr.; Schwaab, G.; Luboinski, B.; Paoletti, C. Electrochemotherapy, a new antitumor treatment: First clinical trial. C. R. Acad. Sci. III 1991, 313, 613–618. [Google Scholar]
- Mir, L.M.; Devauchelle, P.; Quintin-Colonna, F.; Delisle, F.; Doliger, S.; Fradelizi, D.; Belehradek, J., Jr.; Orlowski, S. First clinical trial of cat soft-tissue sarcomas treatment by electrochemotherapy. Br. J. Cancer 1997, 76, 1617–1622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campana, L.G.; Edhemovic, I.; Soden, D.; Perrone, A.M.; Scarpa, M.; Campanacci, L.; Cemazar, M.; Valpione, S.; Miklavcic, D.; Mocellin, S.; et al. Electrochemotherapy-Emerging applications technical advances, new indications, combined approaches, and multi-institutional collaboration. Eur. J. Surg. Oncol. 2019, 45, 92–102. [Google Scholar] [CrossRef]
- Spugnini, E.P.; Baldi, A. Electrochemotherapy in veterinary oncology: From rescue to first line therapy. Methods Mol. Biol. 2014, 1121, 247–256. [Google Scholar]
- Spugnini, E.P.; Baldi, F.; Mellone, P.; Feroce, F.; D’Avino, A.; Bonetto, F.; Vincenzi, B.; Citro, G.; Baldi, A. Patterns of tumor response in canine and feline cancer patients treated with electrochemotherapy: Preclinical data for the standardization of this treatment in pets and humans. J. Transl. Med. 2007, 5, 48. [Google Scholar] [CrossRef] [Green Version]
- Spugnini, E.P.; Fanciulli, M.; Citro, G.; Baldi, A. Preclinical models in electrochemotherapy: The role of veterinary patients. Future Oncol. 2012, 8, 829–837. [Google Scholar] [CrossRef]
- Sersa, G.; Ursic, K.; Cemazar, M.; Heller, R.; Bosnjak, M.; Campana, L.G. Biological factors of the tumour response to electrochemotherapy: Review of the evidence and a research roadmap. Eur. J. Surg. Oncol. 2021. [Google Scholar] [CrossRef] [PubMed]
- Kis, E.G.; Baltas, E.; Ocsai, H.; Vass, A.; Nemeth, I.B.; Varga, E.; Olah, J.; Kemeny, L.; Toth-Molnar, E. Electrochemotherapy in the treatment of locally advanced or recurrent eyelid-periocular basal cell carcinomas. Sci. Rep. 2019, 9, 4285. [Google Scholar] [CrossRef] [PubMed]
- Pichi, B.; Pellini, R.; De Virgilio, A.; Spriano, G. Electrochemotherapy: A well-accepted palliative treatment by patients with head and neck tumours. Acta Otorhinolaryngol. Ital. 2018, 38, 181–187. [Google Scholar]
- Markelc, B.; Cemazar, M.; Sersa, G. Effects of Reversible and Irreversible Electroporation on Endothelial Cells and Tissue Blood Flow; Springer: Cham, Switzerland, 2016. [Google Scholar]
- Sersa, G. The state-of-the-art of electrochemotherapy before the ESOPE study; advantages and clinical uses. Eur. J. Cancer Suppl. Eur. J. Cancer Suppl. 2006, 4, 52–59. [Google Scholar] [CrossRef]
- Spugnini, E.P.; Vincenzi, B.; Carocci, F.; Bonichi, C.; Menicagli, F.; Baldi, A. Combination of bleomycin and cisplatin as adjuvant electrochemotherapy protocol for the treatment of incompletely excised feline injection-site sarcomas: A retrospective study. Open Vet. J. 2020, 10, 267–271. [Google Scholar] [CrossRef]
- Martins Taques, M.; Guedert, R.; Moreno, K.; Monte Mor Rangel, M.; Ota Hisayasu Suzuki, D. Adjuvant electrochemotherapy after debulking in canine bone osteosarcoma infiltration. Artif. Organs. 2021, 45, 309–315. [Google Scholar] [CrossRef]
- Probst, U.; Fuhrmann, I.; Beyer, L.; Wiggermann, P. Electrochemotherapy as a New Modality in Interventional Oncology: A Review. Technol. Cancer Res. Treat. 2018, 17, 1533033818785329. [Google Scholar] [CrossRef] [Green Version]
- Kos, B. Treatment Planning for Electrochemotherapy and Irreversible Electroporation of Deep-Seated Tumors; Springer: Cham, Switzerland, 2017; pp. 1–17. [Google Scholar]
- Granata, V.; Fusco, R.; Setola, S.V.; Piccirillo, M.; Leongito, M.; Palaia, R.; Granata, F.; Lastoria, S.; Izzo, F.; Petrillo, A. Early radiological assessment of locally advanced pancreatic cancer treated with electrochemotherapy. World J. Gastroenterol. 2017, 23, 4767–4778. [Google Scholar] [CrossRef]
- Miklavcic, D.; Davalos, R.V. Electrochemotherapy (ECT) and irreversible electroporation (IRE) -advanced techniques for treating deep-seated tumors based on electroporation. Biomed. Eng. Online 2015, 14, I1. [Google Scholar] [CrossRef] [Green Version]
- Valpione, S.; Campana, L.G.; Rastrelli, M.; Sommariva, A.; Vecchiato, A.; Aliberti, C.; Chiarion-Sileni, V.; Rossi, C.R. Phase I/II Study of Electrochemotherapy with Intravenous Bleomycin and Variable Geometry Electric Fields for the Treatment of Deep and Large Soft Tissue Tumors. Ann. Oncol. Ann. Oncol. 2014, 25, iv510. [Google Scholar] [CrossRef]
- Spugnini, E.P.; Citro, G.; Baldi, A. Adjuvant electrochemotherapy in veterinary patients: A model for the planning of future therapies in humans. J. Exp. Clin. Cancer Res. 2009, 28, 114. [Google Scholar] [CrossRef] [Green Version]
- Spugnini, E.P.; Fais, S.; Azzarito, T.; Baldi, A. Novel Instruments for the Implementation of Electrochemotherapy Protocols: From Bench Side to Veterinary Clinic. J. Cell Physiol. 2017, 232, 490–495. [Google Scholar] [CrossRef]
- Sersa, G.; Cemazar, M.; Miklavcic, D.; Mir, L.M. Electrochemotherapy: Variable anti-tumor effect on different tumor models. Bioold. Bioelectrochem. Bioenerg. 1994, 35, 23–27. [Google Scholar] [CrossRef]
- Cemazar, M.; Sersa, G. Recent advances in electrochemotherapy. Bioelectricity 2019, 1, 204–213. [Google Scholar] [CrossRef]
- Mandal, R.; Chan, T.A. Personalized Oncology Meets Immunology: The Path toward Precision Immunotherapy. Cancer Discov. 2016, 6, 703–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miklavcic, D.; Mali, B.; Kos, B.; Heller, R.; Sersa, G. Electrochemotherapy: From the drawing board into medical practice. Biomed. Eng. Online 2014, 13, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sedlar, A.; Dolinsek, T.; Markelc, B.; Prosen, L.; Kranjc, S.; Bosnjak, M.; Blagus, T.; Cemazar, M.; Sersa, G. Potentiation of electrochemotherapy by intramuscular IL-12 gene electrotransfer in murine sarcoma and carcinoma with different immunogenicity. Radiol. Oncol. 2012, 46, 302–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campana, L.G.; Mocellin, S.; Basso, M.; Puccetti, O.; De Salvo, G.L.; Chiarion-Sileni, V.; Vecchiato, A.; Corti, L.; Rossi, C.R.; Nitti, D. Bleomycin-based electrochemotherapy: Clinical outcome from a single institution’s experience with 52 patients. Ann. Surg. Oncol. 2009, 16, 191–199. [Google Scholar] [CrossRef]
- Campana, L.G.; Testori, A.; Curatolo, P.; Quaglino, P.; Mocellin, S.; Framarini, M.; Borgognoni, L.; Ascierto, P.A.; Mozzillo, N.; Guida, M.; et al. Treatment efficacy with electrochemotherapy: A multi-institutional prospective observational study on 376 patients with superficial tumors. Eur. J. Surg. Oncol. 2016, 42, 1914–1923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esmaeili, N.; Friebe, M. Electrochemotherapy: A Review of Current Status, Alternative IGP Approaches, and Future Perspectives. J. Healthc. Eng. 2019, 2019, 2784516. [Google Scholar] [CrossRef]
- Sersa, G.; Miklavcic, D.; Cemazar, M.; Rudolf, Z.; Pucihar, G.; Snoj, M. Electrochemotherapy in treatment of tumours. Eur. J. Surg. Oncol. 2008, 34, 232–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kos, S.; Blagus, T.; Cemazar, M.; Lampreht Tratar, U.; Stimac, M.; Prosen, L.; Dolinsek, T.; Kamensek, U.; Kranjc, S.; Steinstraesser, L.; et al. Electrotransfer parameters as a tool for controlled and targeted gene expression in skin. Mol. Ther. Nucleic. Acids 2016, 5, e356. [Google Scholar] [CrossRef] [Green Version]
- Gothelf, A.; Mahmood, F.; Dagnaes-Hansen, F.; Gehl, J. Efficacy of transgene expression in porcine skin as a function of electrode choice. Bioelectrochemistry 2011, 82, 95–102. [Google Scholar] [CrossRef] [PubMed]
- Daugimont, L.; Baron, N.; Vandermeulen, G.; Pavselj, N.; Miklavcic, D.; Jullien, M.C.; Cabodevila, G.; Mir, L.M.; Preat, V. Hollow microneedle arrays for intradermal drug delivery and DNA electroporation. J. Membr. Biol. 2010, 236, 117–125. [Google Scholar] [CrossRef]
- Heller, L.C.; Jaroszeski, M.J.; Coppola, D.; McCray, A.N.; Hickey, J.; Heller, R. Optimization of cutaneous electrically mediated plasmid DNA delivery using novel electrode. Gene Ther. 2007, 14, 275–280. [Google Scholar] [CrossRef] [PubMed]
- Heller, L.C.; Heller, R. Electroporation gene therapy preclinical and clinical trials for melanoma. Curr. Gene Ther. 2010, 10, 312–317. [Google Scholar] [CrossRef] [PubMed]
- Calvet, C.Y.; Thalmensi, J.; Liard, C.; Pliquet, E.; Bestetti, T.; Huet, T.; Langlade-Demoyen, P.; Mir, L.M. Optimization of a gene electrotransfer procedure for efficient intradermal immunization with an hTERT-based DNA vaccine in mice. Mol. Ther. Methods Clin. Dev. 2014, 1, 14045. [Google Scholar] [CrossRef]
- Sieni, E.; Dettin, M.; De Robertis, M.; Bazzolo, B.; Conconi, M.T.; Zamuner, A.; Marino, R.; Keller, F.; Campana, L.G.; Signori, E. The Efficiency of Gene Electrotransfer in Breast-Cancer Cell Lines Cultured on a Novel Collagen-Free 3D Scaffold. Cancers 2020, 12, 1043. [Google Scholar] [CrossRef]
- Peri, D.; Deville, M.; Poignard, C.; Signori, E.; Natalini, R. Numerical optimization of plasmid DNA delivery combined with hyaluronidase injection for electroporation protocol. Comput. Methods Programs Biomed. 2020, 186, 105204. [Google Scholar] [CrossRef] [Green Version]
- De Robertis, M.; Pasquet, L.; Loiacono, L.; Bellard, E.; Messina, L.; Vaccaro, S.; Di Pasquale, R.; Fazio, V.M.; Rols, M.P.; Teissie, J.; et al. In Vivo Evaluation of a New Recombinant Hyaluronidase to Improve Gene Electro-Transfer Protocols for DNA-Based Drug Delivery against Cancer. Cancers 2018, 10, 405. [Google Scholar] [CrossRef] [Green Version]
- Pavselj, N.; Miklavcic, D. Numerical modeling in electroporation-based biomedical applications. Radiol. Oncol. 2008, 42, 159–168. [Google Scholar] [CrossRef] [Green Version]
- Kranjc, M.; Miklavcic, D. Electric Field Distribution and Electroporation Threshold; Springer: Cham, Switzerland, 2016. [Google Scholar]
- Ciria, H.M.; Gonzalez, M.M.; Zamora, L.O.; Cabrales, L.E.; Sierra Gonzalez, G.V.; de Oliveira, L.O.; Zanella, R.; Buzaid, A.C.; Parise, O.; Brito, L.M.; et al. Antitumor effects of electrochemical treatment. Chin. J. Cancer Res. 2013, 25, 223–234. [Google Scholar]
- Todorovic, V.; Kamensek, U.; Sersa, G.; Cemazar, M. Changing electrode orientation, but not pulse polarity, increases the efficacy of gene electrotransfer to tumors in vivo. Bioelectrochemistry 2014, 100, 119–127. [Google Scholar] [CrossRef]
- Venereau, E.; Ceriotti, C.; Bianchi, M.E. DAMPs from Cell Death to New Life. Front. Immunol. 2015, 6, 422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pradeu, T.; Cooper, E.L. The danger theory: 20 years later. Front. Immunol. 2012, 3, 287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 1994, 12, 991–1045. [Google Scholar] [CrossRef]
- Kono, H.; Rock, K.L. How dying cells alert the immune system to danger. Nat. Rev. Immunol. 2008, 8, 279–289. [Google Scholar] [CrossRef] [PubMed]
- Haen, S.P.; Loffler, M.W.; Rammensee, H.G.; Brossart, P. Towards new horizons: Characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 2020, 17, 595–610. [Google Scholar] [CrossRef] [PubMed]
- Zamora, A.E.; Crawford, J.C.; Thomas, P.G. Hitting the Target: How T Cells Detect and Eliminate Tumors. J. Immunol. 2018, 200, 392–399. [Google Scholar] [CrossRef]
- Calvet, C.Y.; Famin, D.; Andre, F.M.; Mir, L.M. Electrochemotherapy with bleomycin induces hallmarks of immunogenic cell death in murine colon cancer cells. Oncoimmunology 2014, 3, e28131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roux, S.; Bernat, C.; Al-Sakere, B.; Ghiringhelli, F.; Opolon, P.; Carpentier, A.F.; Zitvogel, L.; Mir, L.M.; Robert, C. Tumor destruction using electrochemotherapy followed by CpG oligodeoxynucleotide injection induces distant tumor responses. Cancer Immunol. Immunother. 2008, 57, 1291–1300. [Google Scholar] [CrossRef]
- Heath, W.R.; Belz, G.T.; Behrens, G.M.; Smith, C.M.; Forehan, S.P.; Parish, I.A.; Davey, G.M.; Wilson, N.S.; Carbone, F.R.; Villadangos, J.A. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol. Rev. 2004, 199, 9–26. [Google Scholar] [CrossRef]
- Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef]
- Jakstys, B.; Jakutaviciute, M.; Uzdavinyte, D.; Satkauskiene, I.; Satkauskas, S. Correlation between the loss of intracellular molecules and cell viability after cell electroporation. Bioelectrochemistry 2020, 135, 107550. [Google Scholar] [CrossRef]
- Schultheis, K.; Smith, T.R.F.; Kiosses, W.B.; Kraynyak, K.A.; Wong, A.; Oh, J.; Broderick, K.E. Delineating the Cellular Mechanisms Associated with Skin Electroporation. Hum. Gene Ther. Methods 2018, 29, 177–188. [Google Scholar] [CrossRef] [PubMed]
- Zhan, Y.; Sun, C.; Cao, Z.; Bao, N.; Xing, J.; Lu, C. Release of intracellular proteins by electroporation with preserved cell viability. Anal. Chem. 2012, 84, 8102–8105. [Google Scholar] [CrossRef] [PubMed]
- Galluzzi, L.; Vitale, I.; Warren, S.; Adjemian, S.; Agostinis, P.; Martinez, A.B.; Chan, T.A.; Coukos, G.; Demaria, S.; Deutsch, E.; et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [Green Version]
- Mlakar, V.; Todorovic, V.; Cemazar, M.; Glavac, D.; Sersa, G. Electric pulses used in electrochemotherapy and electrogene therapy do not significantly change the expression profile of genes involved in the development of cancer in malignant melanoma cells. BMC Cancer 2009, 9, 299. [Google Scholar] [CrossRef] [Green Version]
- Panaretakis, T.; Kepp, O.; Brockmeier, U.; Tesniere, A.; Bjorklund, A.C.; Chapman, D.C.; Durchschlag, M.; Joza, N.; Pierron, G.; van Endert, P.; et al. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. EMBO J. 2009, 28, 578–590. [Google Scholar] [CrossRef]
- Gabriel, B.; Teissie, J. Generation of reactive-oxygen species induced by electropermeabilization of Chinese hamster ovary cells and their consequence on cell viability. Eur. J. Biochem. 1994, 223, 25–33. [Google Scholar] [CrossRef] [PubMed]
- Di Gennaro, P.; Gerlini, G.; Urso, C.; Sestini, S.; Brandani, P.; Pimpinelli, N.; Borgognoni, L. CD4(+)FOXP3(+) T regulatory cells decrease and CD3(+)CD8(+) T cells recruitment in TILs from melanoma metastases after electrochemotherapy. Clin. Exp. Metastasis 2016, 33, 787–798. [Google Scholar] [CrossRef]
- Andersson, U.; Wang, H.; Palmblad, K.; Aveberger, A.C.; Bloom, O.; Erlandsson-Harris, H.; Janson, A.; Kokkola, R.; Zhang, M.; Yang, H.; et al. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J. Exp. Med. 2000, 192, 565–570. [Google Scholar] [CrossRef]
- Peng, B.; Zhao, Y.; Xu, L.; Xu, Y. Electric pulses applied prior to intramuscular DNA vaccination greatly improve the vaccine immunogenicity. Vaccine 2007, 25, 2064–2073. [Google Scholar] [CrossRef]
- Prevc, A.; Bedina Zavec, A.; Cemazar, M.; Kloboves-Prevodnik, V.; Stimac, M.; Todorovic, V.; Strojan, P.; Sersa, G. Bystander Effect Induced by Electroporation is Possibly Mediated by Microvesicles and Dependent on Pulse Amplitude, Repetition Frequency and Cell Type. J. Membr. Biol. 2016, 249, 703–711. [Google Scholar] [CrossRef]
- Sersa, G.; Miklavcic, D.; Cemazar, M.; Belehradek, J.; Jarm, T.; Mir, L.M. Electrochemotherapy with CDDP on LPB sarcoma: Comparison of the anti-tumor effectiveness in immunocompotent and immunodeficient mice. Bioelectrochem. Bioenerg. Bioelectrochem. Bioenerg. 1997, 43, 279–283. [Google Scholar] [CrossRef]
- Belizario, J.E. Immunodeficient Mouse Models: An Overview. TOIJ Open Immunol. J. 2009, 2, 79–85. [Google Scholar] [CrossRef]
- Kepp, O.; Marabelle, A.; Zitvogel, L.; Kroemer, G. Oncolysis without viruses-inducing systemic anticancer immune responses with local therapies. Nat. Rev. Clin. Oncol. 2020, 17, 49–64. [Google Scholar] [CrossRef] [PubMed]
- Sersa, G.; Teissie, J.; Cemazar, M.; Signori, E.; Kamensek, U.; Marshall, G.; Miklavcic, D. Electrochemotherapy of tumors as in situ vaccination boosted by immunogene electrotransfer. Cancer Immunol. Immunother. 2015, 64, 1315–1327. [Google Scholar] [CrossRef]
- Gerlini, G.; Di Gennaro, P.; Borgognoni, L. Enhancing anti-melanoma immunity by electrochemotherapy and in vivo dendritic-cell activation. Oncoimmunology 2012, 1, 1655–1657. [Google Scholar] [CrossRef] [Green Version]
- Andersen, M.H.; Gehl, J.; Reker, S.; Pedersen, L.O.; Becker, J.C.; Geertsen, P.; thor Straten, P. Dynamic changes of specific T cell responses to melanoma correlate with IL-2 administration. Semin. Cancer Biol. 2003, 13, 449–459. [Google Scholar] [CrossRef]
- Ginn, S.L.; Amaya, A.K.; Alexander, I.E.; Edelstein, M.; Abedi, M.R. Gene therapy clinical trials worldwide to 2017: An update. J. Gene Med. 2018, 20, e3015. [Google Scholar] [CrossRef] [PubMed]
- Abedia. Available online: https://a873679.fmphost.com/fmi/webd/GTCT (accessed on 13 February 2021).
- Chiarella, P.; Fazio, V.M.; Signori, E. Electroporation in DNA vaccination protocols against cancer. Curr. Drug Metab. 2013, 14, 291–299. [Google Scholar] [CrossRef]
- Chiarella, P.; Massi, E.; De Robertis, M.; Fazio, V.M.; Signori, E. Strategies for effective naked-DNA vaccination against infectious diseases. Recent Pat. Antiinfect Drug Discov. 2008, 3, 93–101. [Google Scholar] [CrossRef] [PubMed]
- Aihara, H.; Miyazaki, J. Gene transfer into muscle by electroporation in vivo. Nat. Biotechnol. 1998, 16, 867–870. [Google Scholar] [CrossRef] [PubMed]
- Heller, L.C. Principles of Electroporation for Gene Therapy; Springer: Cham, Switzerland, 2017. [Google Scholar]
- Chiarella, P.; Massi, E.; De Robertis, M.; Sibilio, A.; Parrella, P.; Fazio, V.M.; Signori, E. Electroporation of skeletal muscle induces danger signal release and antigen-presenting cell recruitment independently of DNA vaccine administration. Expert Opin. Biol. Ther. 2008, 8, 1645–1657. [Google Scholar] [CrossRef] [PubMed]
- Mukhopadhyay, A.; Wright, J.; Shirley, S.; Canton, D.A.; Burkart, C.; Connolly, R.J.; Campbell, J.S.; Pierce, R.H. Characterization of abscopal effects of intratumoral electroporation-mediated IL-12 gene therapy. Gene Ther. 2019, 26, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Shirley, S.A.; Lundberg, C.G.; Li, F.; Burcus, N.; Heller, R. Controlled gene delivery can enhance therapeutic outcome for cancer immune therapy for melanoma. Curr. Gene Ther. 2015, 15, 32–43. [Google Scholar] [CrossRef] [PubMed]
- ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/results?term=electroporation+NOT+irreversible+NOT+calcium+NOT+infection&recrs=a (accessed on 23 February 2021).
- AAHSD–AVMA (American Veterinary Medical Association) Animal Health Studies Database. Available online: https://ebusiness.avma.org/aahsd/study_search.aspx (accessed on 13 February 2021).
- Kamensek, U.; Tesic, N.; Sersa, G.; Cemazar, M. Clinically Usable Interleukin 12 Plasmid without an Antibiotic Resistance Gene: Functionality and Toxicity Study in Murine Melanoma Model. Cancers 2018, 10, 60. [Google Scholar] [CrossRef] [Green Version]
- Lasek, W.; Zagozdzon, R.; Jakobisiak, M. Interleukin 12: Still a promising candidate for tumor immunotherapy? Cancer Immunol. Immunother. 2014, 63, 419–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, M.; Mizoguchi, I.; Morishima, N.; Chiba, Y.; Mizuguchi, J.; Yoshimoto, T. Regulation of antitumor immune responses by the IL-12 family cytokines, IL-12, IL-23, and IL-27. Clin. Dev. Immunol. 2010, 2010. [Google Scholar] [CrossRef] [Green Version]
- Algazi, A.P.; Twitty, C.G.; Tsai, K.K.; Le, M.; Pierce, R.; Browning, E.; Hermiz, R.; Canton, D.A.; Bannavong, D.; Oglesby, A.; et al. Phase II Trial of IL-12 Plasmid Transfection and PD-1 Blockade in Immunologically Quiescent Melanoma. Clin. Cancer Res. 2020, 26, 2827–2837. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.N.; Sun, E.G.; Cho, S.H. IL-12 Enhances Immune Response by Modulation of Myeloid Derived Suppressor Cells in Tumor Microenvironment. Chonnam. Med. J. 2019, 55, 31–39. [Google Scholar] [CrossRef] [Green Version]
- Zhao, J.; Zhao, J.; Perlman, S. Differential effects of IL-12 on Tregs and non-Treg T cells: Roles of IFN-gamma, IL-2 and IL-2R. PLoS ONE 2012, 7, e46241. [Google Scholar] [CrossRef] [PubMed]
- Steding, C.E.; Wu, S.T.; Zhang, Y.; Jeng, M.H.; Elzey, B.D.; Kao, C. The role of interleukin-12 on modulating myeloid-derived suppressor cells, increasing overall survival and reducing metastasis. Immunology 2011, 133, 221–238. [Google Scholar] [CrossRef]
- Feng, T.; Cao, A.T.; Weaver, C.T.; Elson, C.O.; Cong, Y. Interleukin-12 converts Foxp3+ regulatory T cells to interferon-gamma-producing Foxp3+ T cells that inhibit colitis. Gastroenterology 2011, 140, 2031–2043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Henry, C.J.; Ornelles, D.A.; Mitchell, L.M.; Brzoza-Lewis, K.L.; Hiltbold, E.M. IL-12 produced by dendritic cells augments CD8+ T cell activation through the production of the chemokines CCL1 and CCL17. J. Immunol. 2008, 181, 8576–8584. [Google Scholar] [CrossRef] [Green Version]
- Dunn, G.P.; Ikeda, H.; Bruce, A.T.; Koebel, C.; Uppaluri, R.; Bui, J.; Chan, R.; Diamond, M.; White, J.M.; Sheehan, K.C.; et al. Interferon-gamma and cancer immunoediting. Immunol. Res. 2005, 32, 231–245. [Google Scholar] [CrossRef]
- Yue, F.Y.; Geertsen, R.; Hemmi, S.; Burg, G.; Pavlovic, J.; Laine, E.; Dummer, R. IL-12 directly up-regulates the expression of HLA class I, HLA class II and ICAM-1 on human melanoma cells: A mechanism for its antitumor activity? Eur. J. Immunol. 1999, 29, 1762–1773. [Google Scholar] [CrossRef]
- Shi, G.; Edelblute, C.; Arpag, S.; Lundberg, C.; Heller, R. IL-12 Gene Electrotransfer Triggers a Change in Immune Response within Mouse Tumors. Cancers 2018, 10, 498. [Google Scholar] [CrossRef] [Green Version]
- Lampreht Tratar, U.; Loiacono, L.; Cemazar, M.; Kamensek, U.; Fazio, V.M.; Sersa, G.; Signori, E. Gene Electrotransfer of Plasmid-Encoding IL-12 Recruits the M1 Macrophages and Antigen-Presenting Cells Inducing the Eradication of Aggressive B16F10 Murine Melanoma. Mediat. Inflamm. 2017, 2017, 5285890. [Google Scholar] [CrossRef]
- Herndler-Brandstetter, D.; Ishigame, H.; Shinnakasu, R.; Plajer, V.; Stecher, C.; Zhao, J.; Lietzenmayer, M.; Kroehling, L.; Takumi, A.; Kometani, K.; et al. KLRG1(+) Effector CD8(+) T Cells Lose KLRG1, Differentiate into All Memory T Cell Lineages, and Convey Enhanced Protective Immunity. Immunity 2018, 48, 716–729.e8. [Google Scholar] [CrossRef] [Green Version]
- Sin, J.I.; Park, J.B.; Lee, I.H.; Park, D.; Choi, Y.S.; Choe, J.; Celis, E. Intratumoral electroporation of IL-12 cDNA eradicates established melanomas by Trp2(180-188)-specific CD8+ CTLs in a perforin/granzyme-mediated and IFN-gamma-dependent manner: Application of Trp2(180-188) peptides. Cancer Immunol. Immunother. 2012, 61, 1671–1682. [Google Scholar] [CrossRef] [PubMed]
- Shibata, M.A.; Ito, Y.; Morimoto, J.; Kusakabe, K.; Yoshinaka, R.; Otsuki, Y. In vivo electrogene transfer of interleukin-12 inhibits tumor growth and lymph node and lung metastases in mouse mammary carcinomas. J. Gene Med. 2006, 8, 335–352. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Zhang, L.; Torrero, M.; Cannon, M.; Barret, R. Administration route- and immune cell activation-dependent tumor eradication by IL12 electrotransfer. Mol. Ther. 2005, 12, 942–949. [Google Scholar] [CrossRef] [PubMed]
- Cemazar, M.; Ambrozic Avgustin, J.; Pavlin, D.; Sersa, G.; Poli, A.; Krhac Levacic, A.; Tesic, N.; Lampreht Tratar, U.; Rak, M.; Tozon, N. Efficacy and safety of electrochemotherapy combined with peritumoral IL-12 gene electrotransfer of canine mast cell tumours. Vet. Comp. Oncol. 2017, 15, 641–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pavlin, D.; Cemazar, M.; Sersa, G.; Tozon, N. IL-12 based gene therapy in veterinary medicine. J. Transl. Med. 2012, 10, 234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reed, S.D.; Fulmer, A.; Buckholz, J.; Zhang, B.; Cutrera, J.; Shiomitsu, K.; Li, S. Bleomycin/interleukin-12 electrochemogene therapy for treating naturally occurring spontaneous neoplasms in dogs. Cancer Gene Ther. 2010, 17, 457–464. [Google Scholar] [CrossRef] [Green Version]
- Kishida, T.; Asada, H.; Itokawa, Y.; Yasutomi, K.; Shin-Ya, M.; Gojo, S.; Cui, F.D.; Ueda, Y.; Yamagishi, H.; Imanishi, J.; et al. Electrochemo-gene therapy of cancer: Intratumoral delivery of interleukin-12 gene and bleomycin synergistically induced therapeutic immunity and suppressed subcutaneous and metastatic melanomas in mice. Mol. Ther. 2003, 8, 738–745. [Google Scholar] [CrossRef] [PubMed]
- Milevoj, N.; Tratar, U.L.; Nemec, A.; Brozic, A.; Znidar, K.; Sersa, G.; Cemazar, M.; Tozon, N. A combination of electrochemotherapy, gene electrotransfer of plasmid encoding canine IL-12 and cytoreductive surgery in the treatment of canine oral malignant melanoma. Res. Vet. Sci. 2019, 122, 40–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef] [Green Version]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
- Brizio, M.; Fava, P.; Astrua, C.; Cavaliere, G.; Savoia, P. Complete regression of melanoma skin metastases after electrochemotherapy plus ipilimumab treatment: An unusual clinical presentation. Eur. J. Dermatol. 2015, 25, 271–272. [Google Scholar] [CrossRef]
- Mozzillo, N.; Simeone, E.; Benedetto, L.; Curvietto, M.; Giannarelli, D.; Gentilcore, G.; Camerlingo, R.; Capone, M.; Madonna, G.; Festino, L.; et al. Assessing a novel immuno-oncology-based combination therapy: Ipilimumab plus electrochemotherapy. Oncoimmunology 2015, 4, e1008842. [Google Scholar] [CrossRef] [PubMed]
- Karaca, B.; Yayla, G.; Erdem, M.; Gurler, T. Electrochemotherapy with anti-PD-1 treatment induced durable complete response in heavily pretreated metastatic melanoma patient. Anticancer Drugs 2018, 29, 190–196. [Google Scholar] [CrossRef]
- Heppt, M.V.; Eigentler, T.K.; Kahler, K.C.; Herbst, R.A.; Goppner, D.; Gambichler, T.; Ulrich, J.; Dippel, E.; Loquai, C.; Schell, B.; et al. Immune checkpoint blockade with concurrent electrochemotherapy in advanced melanoma: A retrospective multicenter analysis. Cancer Immunol. Immunother. 2016, 65, 951–959. [Google Scholar] [CrossRef] [PubMed]
- Kos, S.; Lopes, A.; Preat, V.; Cemazar, M.; Lampreht Tratar, U.; Ucakar, B.; Vanvarenberg, K.; Sersa, G.; Vandermeulen, G. Intradermal DNA vaccination combined with dual CTLA-4 and PD-1 blockade provides robust tumor immunity in murine melanoma. PLoS ONE 2019, 14, e0217762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamolinara, A.; Stramucci, L.; Hysi, A.; Iezzi, M.; Marchini, C.; Mariotti, M.; Amici, A.; Curcio, C. Intradermal DNA Electroporation Induces Cellular and Humoral Immune Response and Confers Protection against HER2/neu Tumor. J. Immunol. Res. 2015, 2015, 159145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mali, B.; Jarm, T.; Snoj, M.; Sersa, G.; Miklavcic, D. Antitumor effectiveness of electrochemotherapy: A systematic review and meta-analysis. Eur. J. Surg. Oncol 2013, 39, 4–16. [Google Scholar] [CrossRef] [PubMed]
- Gerlini, G.; Sestini, S.; Di Gennaro, P.; Urso, C.; Pimpinelli, N.; Borgognoni, L. Dendritic cells recruitment in melanoma metastasis treated by electrochemotherapy. Clin. Exp. Metastasis 2013, 30, 37–45. [Google Scholar] [CrossRef] [PubMed]
- Quaglino, P.; Osella-Abate, S.; Marenco, F.; Nardo, T.; Gado, C.; Novelli, M.; Savoia, P.; Bernengo, M.G. FoxP3 expression on melanoma cells is related to early visceral spreading in melanoma patients treated by electrochemotherapy. Pigment. Cell Melanoma Res. 2011, 24, 734–736. [Google Scholar] [CrossRef]
- Ursic, K.; Kos, S.; Kamensek, U.; Cemazar, M.; Miceska, S.; Markelc, B.; Bucek, S.; Staresinic, B.; Kloboves Prevodnik, V.; Heller, R.; et al. Potentiation of electrochemotherapy effectiveness by immunostimulation with IL-12 gene electrotransfer in mice is dependent on tumor immune status. J. Control. Release 2021, 332, 623–635. [Google Scholar] [CrossRef]
- Lucas, M.L.; Heller, L.; Coppola, D.; Heller, R. IL-12 plasmid delivery by in vivo electroporation for the successful treatment of established subcutaneous B16.F10 melanoma. Mol. Ther. 2002, 5, 668–675. [Google Scholar] [CrossRef]
- Pavlin, D.; Cemazar, M.; Kamensek, U.; Tozon, N.; Pogacnik, A.; Sersa, G. Local and systemic antitumor effect of intratumoral and peritumoral IL-12 electrogene therapy on murine sarcoma. Cancer Biol. Ther. 2009, 8, 2114–2122. [Google Scholar] [CrossRef]
- Bhatia, S.; Longino, N.V.; Miller, N.J.; Kulikauskas, R.; Iyer, J.G.; Ibrani, D.; Blom, A.; Byrd, D.R.; Parvathaneni, U.; Twitty, C.G.; et al. Intratumoral Delivery of Plasmid IL12 Via Electroporation Leads to Regression of Injected and Noninjected Tumors in Merkel Cell Carcinoma. Clin. Cancer Res. 2020, 26, 598–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maglietti, F.; Tellado, M.; Olaiz, N.; Michinski, S.; Marshall, G. Combined local and systemic bleomycin administration in electrochemotherapy to reduce the number of treatment sessions. Radiol. Oncol. 2016, 50, 58–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Groselj, A.; Kranjc, S.; Bosnjak, M.; Krzan, M.; Kosjek, T.; Prevc, A.; Cemazar, M.; Sersa, G. Vascularization of the tumours affects the pharmacokinetics of bleomycin and the effectiveness of electrochemotherapy. Basic Clin. Pharmacol. Toxicol. 2018, 123, 247–256. [Google Scholar] [CrossRef] [PubMed]
- De Virgilio, A.; Ralli, M.; Longo, L.; Mancini, P.; Attanasio, G.; Atturo, F.; De Vincentiis, M.; Greco, A. Electrochemotherapy in head and neck cancer: A review of an emerging cancer treatment. Oncol. Lett. 2018, 16, 3415–3423. [Google Scholar] [CrossRef] [Green Version]
- Campana, L.G.; Clover, A.J.; Valpione, S.; Quaglino, P.; Gehl, J.; Kunte, C.; Snoj, M.; Cemazar, M.; Rossi, C.R.; Miklavcic, D.; et al. Recommendations for improving the quality of reporting clinical electrochemotherapy studies based on qualitative systematic review. Radiol. Oncol. 2016, 50, 1–13. [Google Scholar] [CrossRef]
Research Aim | Neoplasms | Clinical Trial | Reference Centers |
---|---|---|---|
VGX-3100 and electroporation in treating patients with HIV positive high-grade anal lesions |
| NCT03603808 |
|
Tavo with E electroporation, pembrolizumab and epacadostat in patients with unresectable head and neck cancer |
| NCT03823131 |
|
Neoadjuvant immunotherapy with Tavo + electroporation in combination with Nivo. in melanoma patients |
| NCT04526730 |
|
REVEAL 2 Trial (Evaluation of VGX-3100 and Electroporation for the treatment of cervical HSIL) |
| NCT03721978 |
|
HPV DNA vaccine via electroporation for HPV16 positive cervical neoplasia |
| NCT04131413 |
|
Tavo and pembrolizumab in patients with stage III/IV melanoma progressing on pembrolizumab or nivolumab treatment (Keynote 695) |
| NCT03132675 |
|
Tavo and pembrolizumab with or without chemotherapy in patients with inoperable locally advanced or metastatic TNBC |
| NCT03567720 |
|
Electrochemotherapy for noncurable gastric cancer |
| NCT04139070 |
|
A study of JNJ-64300535 in healthy participants |
| NCT04736147 |
|
INO 5401 vaccination in BRCA1/2 mutation carriers |
| NCT04367675 |
|
DNA plasmid-encoding interleukin-12/HPV DNA plasmids therapeutic vaccine INO-3112 and durvalumab in treating patients with recurrent or metastatic human papillomavirus associated cancers |
| NCT03439085 |
|
Neoantigen DNA vaccine alone vs. neoantigen DNA vaccine plus durvalumab in triple negative breast cancer patients following standard of care therapy |
| NCT03199040 |
|
A study evaluating UCART019 in patients with relapsed or refractory CD19+ leukemia and lymphoma |
| NCT03166878 |
|
GNOS-PV02 personalized neoantigen vaccine, INO-9012 and pembrolizumab in subjects with advanced HCC |
| NCT04251117 |
|
Safety and immune response to a mammaglobin-A DNA vaccine in breast cancer patients undergoing neoadjuvant endocrine therapy |
| NCT02204098 |
|
Platform study for prostate researching translational endpoints correlated to response to inform use of novelcombinations |
| NCT03835533 |
|
Electrochemotherapy of gynecological cancers |
| NCT04760327 |
|
EndoVE endoscopic treatment for esophageal and gastric cancer |
| NCT04649372 |
|
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Luz, J.C.d.S.d.; Antunes, F.; Clavijo-Salomon, M.A.; Signori, E.; Tessarollo, N.G.; Strauss, B.E. Clinical Applications and Immunological Aspects of Electroporation-Based Therapies. Vaccines 2021, 9, 727. https://doi.org/10.3390/vaccines9070727
Luz JCdSd, Antunes F, Clavijo-Salomon MA, Signori E, Tessarollo NG, Strauss BE. Clinical Applications and Immunological Aspects of Electroporation-Based Therapies. Vaccines. 2021; 9(7):727. https://doi.org/10.3390/vaccines9070727
Chicago/Turabian StyleLuz, Jean Carlos dos Santos da, Fernanda Antunes, Maria Alejandra Clavijo-Salomon, Emanuela Signori, Nayara Gusmão Tessarollo, and Bryan E. Strauss. 2021. "Clinical Applications and Immunological Aspects of Electroporation-Based Therapies" Vaccines 9, no. 7: 727. https://doi.org/10.3390/vaccines9070727
APA StyleLuz, J. C. d. S. d., Antunes, F., Clavijo-Salomon, M. A., Signori, E., Tessarollo, N. G., & Strauss, B. E. (2021). Clinical Applications and Immunological Aspects of Electroporation-Based Therapies. Vaccines, 9(7), 727. https://doi.org/10.3390/vaccines9070727