Preclinical Cold Atmospheric Plasma Cancer Treatment: Comparison
Please note this is a comparison between Version 2 by Rita Xu and Version 1 by Dayun Yan.

Cold atmospheric plasma (CAP) is generated in a rapid yet low-energy input streamer-discharge process under atmospheric pressure conditions and environment. CAP is an ionized gas with a low ionization degree and plenty of reactive species and radicals. These reactive components, and their near-room temperature nature, make CAP a powerful tool in medical applications, particularly cancer therapy. Here, wresearchers systematically introduced the research status of the preclinical application of CAP in cancer therapy, particularly in vivo studies. 

  • cold atmospheric plasma
  • cancer treatment
  • anti-tumor therapy

1. CAP and Plasma Sources

CAP has been widely used in several branches of medicine, including wound healing, microorganism sterilization, biofilm inactivation, and cancer therapy [1][2][3]. CAP is an ionized gas composed of reactive compounds such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) [4][5], and is designed to work under atmospheric pressure at near room temperature [6]. CAP sources, such as dielectric barrier discharge (DBD), atmospheric pressure plasma jet (APPJ), and plasma torch, are the foundation for plasma biomedical applications [7][8]. Six of the most commonly used CAP sources are shown in Figure 1. Alternating current (AC) is a typical power supply for these CAP sources [9]
Figure 1. Typical CAP sources used in plasma medicine. Type a. Volume DBD. Type b. Surface DBD. Type c. Two-ring electrodes APPJ. Type d. Plasma torch using ring electrode. Type e. One central electrode-one ring electrode APPJ. Type f. Plasma torch using a central electrode.

2. General Picture of In Vitro Studies

To date, the promising anti-cancer performance of CAP treatment in vitro has been extensively demonstrated in dozens of cancer types, including skin, breast, colorectal, brain, lung, cervical, head and neck cancer [3][10]. Plenty of reviews and articles have been published, most of them focused on in vitro studies and corresponding conclusions [11][12][13]. Several basic cellular responses have been repeatedly observed in the publications and are listed in Table 1. These basic cellular responses build the foundation for understanding the anti-cancer effect of CAP treatment in vitro and address some in vivo observations.
Table 1. Basic cancer cellular responses of CAP treatment in vitro.
Like most medical studies, the conclusions obtained from in vitro studies cannot be easily used to directly predict the performance of in vivo studies. For example, the relatively dry skin barrier between plasma and targeted cancerous tissues or cells under the skin is quite different from the commonly accepted experimental conditions in vitro. The in vitro environment mainly involves a relatively thick medium layer to facilitate the transition of some short-lived reactive species in the gas phase to long-lived reactive species in the liquid phase. Moreover, both long-lived and short-lived reactive species will have complex reactions at this gas/liquid interface. In vivo studies play a cornerstone role in plasma medicine before CAP can be used in clinical therapy [40][41][42]. More importantly, in vivo studies directly assess the CAP treatment’s safety on tissues and animals, such as carcinogenicity [43]. In thiRes review, ourearchers preclinical studies’ discussion will be just limited to in vivo studies.
Compared to the abundant in vitro investigations, in vivo studies have gradually become the main approach to discovering novel tissue responses to CAP treatment. Animal models’ design directly determines the use of CAP in the in vivo studies. So far, three types of animal models have been widely used to demonstrate the anti-tumor efficacy of CAP treatment: subcutaneous model, intraperitoneal model, and orthotopic model [44]. To date, most of CAP’s anti-tumor capability was demonstrated using subcutaneous models.
Subcutaneous models provided the earliest and the most apparent demonstration for the feasibility of using CAP as an anti-tumor modality. The earliest in vivo works were demonstrated by Marc Vandamme, et al. and Michael Keidar, et al. between 2010–2011. They used a glioblastoma U87MG xenograft mouse model and bladder xenograft tumor model to test a CAP treatment’s in vivo efficacy for just a few minutes, respectively. The two pioneering research articles demonstrated a drastic tumor volume reduction of more than 50% after floating electrode DBD treatment and APPJ treatment [41][45]. Correspondingly, the survival length of mice strongly increased by more than 60% in the two models [41][45]. These two works also first tested the safety of using CAP in animal studies. Results showed no toxic side effects or potential physical damage from plasma.
 
Due to the subcutaneous nature of melanomas, it has become one of the more promising candidates for CAP-based cancer therapy. Many studies have been performed on melanoma models [7]. A nanosecond pulsed DBD (nsP DBD) completely eradicated the xenografted melanoma tumor in mice after direct treatment on the skin above the melanoma. Histology of an nsP DBD treated tumor showed a typical red skin staining without tumor tissue below the epithelium. Correspondingly, the survival rate of mice increased from 0% to 66.7% 20–40 days succeeding the nsp DBD treatment [48].
Similar trends have been repeatedly observed in a series of following studies. Table 2 lists representative in vivo anti-tumor demonstrations (2010–2018) on subcutaneous xenograft tumors in mice. In the subcutaneous model, CAP treatment was mainly carried out by treating the skin above tumorous tissues. In such a setting, the effective factor, either chemical or physical factors in CAP, must penetrate the skin barrier and further trigger biological pathways to inhibit tumorous growth, therefore providing CAP treatment as a potential non-invasive anti-tumor modality. Among these studies, a general trend has been repeatedly observed. That is, a treatment just above the skin could strongly inhibit the growth of tumors and significantly extend the life length of mice [45]
Table 2. Representative in vivo demonstrations on subcutaneous xenografted tumor models (2010–2018).
Ref Years Tumor Types Tumor Size Survival Rate Tumor Diagnostics
[45] 2010 Glioblastoma Decreased N/A Bioluminescence imaging
[41] 2010 Bladder cancer Decreased Increased Tissue size measurement
[46] 2011 Glioblastoma Decreased Increased Bioluminescence imaging
[47] 2012 Pancreatic carcinoma Decreased N/A Bioluminescence imaging
[48] 2012 Glioblastoma Decreased N/A Bioluminescence imaging
[49] 2013 Neuroblastoma Decreased Increased Tissue size measurement
] Mitochondrial Damage 2010
[50] 2014 Melanoma Decreased N/A Tissue size measurement [20]
[51]Rise of Intracellular ROS 2011
2014 Head and neck cancer Decreased N/A Tissue size measurement [21] Chemically-based Sensitization to Drugs 2013
[40] 2015 Melanoma Decreased Increased Tissue size measurement [22] Selective Rise of Intracellular ROS 2013
[52] 2015 Endometrioid adenocarcinoma Decreased N/A Tissue size measurement [23] Senescence 2013
[53] 2016 Glioblastoma Decreased N/A Tissue size measurement [24] Immunogenic Cell Death
[2015
54] 2016 Breast cancer Decreased N/A Tissue size measurement [25] Cell-based H2O
[42]2 Generation 20172017
Melanoma Decreased N/A Bioluminescence imaging [26]
[Autophagy-associated Cell Death 2017
55] 2018 [27] Activation Phenomena 2018
[28] Physically-triggered Necrosis 2020
[29] Pyroptosis 2020
[30] Physically-based Sensitization to Drugs 2021
Together, some general conclusions can be summarized here. (1) Reactive species play a critical role in the liquid phase-based experimental setting [31]. Apoptosis, necrosis, and autophagy are the main cellular death approaches following CAP treatment with an adequately large dose [32]. (2) Physical factors, particularly electromagnetic effects from plasma, may exert a clear impact on cells, such as bacteria and mammalian cells [33][34]. (3) A noticeable rise in intracellular ROS is a pivotal cellular response to CAP treatment, which further triggers downstream cellular damage, including DNA damage, mitochondrial damage, cellular membrane damage, and cell death [1]. (4) Aqueous environment, such as a medium layer, plays a pivotal role in facilitating the transition of short-lived reactive species in the gas phase into long-lived reactive species in the liquid phase [35][36][37]. For in vitro studies, a medium layer is necessary for experimental design and is responsible for most observed cellular responses after CAP treatment, particularly for the cases involving CAP-treated solutions or media [32][38]. (5) CAP shows a selective killing effect on cancer cell lines compared to their counterpart normal cell lines in many cases [39].

3. Direct CAP Treatment In Vivo

Colorectal tumor
Decreased
N/A
Tissue size measurement

References

  1. Von Woedtke, T.; Schmidt, A.; Bekeschus, S.; Wende, K.; Weltmann, K.D. Plasma medicine: A field of applied redox biology. In Vivo 2019, 33, 1011–1026.
  2. Fridman, G.; Friedman, G.; Gutsol, A.; Shekhter, A.B.; Vasilets, V.N.; Fridman, A. Applied plasma medicine. Plasma Processes Polym. 2008, 5, 503–533.
  3. Keidar, M. Plasma for cancer treatment. Plasma Sources Sci. Technol. 2015, 24, 033001.
  4. Keidar, M. A prospectus on innovations in the plasma treatment of cancer. Phys. Plasmas 2018, 25, 083504.
  5. Graves, D.B. Reactive species from cold atmospheric plasma: Implications for cancer therapy. Plasma Processes Polym. 2014, 11, 1120–1127.
  6. Tendero, C.; Tixier, C.; Tristant, P.; Desmaison, J.; Leprince, P. Atmospheric pressure plasmas: A review. Spectrochim. Acta B At. Spectrosc. 2006, 61, 2–30.
  7. Kogelschatz, U. Atmospheric-pressure plasma technology. Plasma Phys. Control. Fusion 2004, 46, B63–B75.
  8. Park, G.Y.; Park, S.J.; Choi, M.Y.; Koo, I.G.; Byun, J.H.; Hong, J.W.; Sim, J.Y.; Collins, G.J.; Lee, J.K. Atmospheric-pressure plasma sources for biomedical applications. Plasma Sources Sci. Technol. 2012, 21, 043001.
  9. Yan, D.; Lin, L.; Xu, W.; Nourmohammadi, N. Cold plasma-based control of the activation of pancreatic adenocarcinoma cells. J. Phys. D Appl. Phys. 2019, 52.44, 445202.
  10. Yan, D.; Sherman, J.H.; Keidar, M. Cold atmospheric plasma, a novel promising anti-cancer treatment modality. Oncotarget 2017, 8, 15977.
  11. Yan, D.; Talbot, A.; Nourmohammadi, N.; Sherman, J.H.; Cheng, X.; Keidar, M. Toward understanding the selective anticancer capacity of cold atmospheric plasma—A model based on aquaporins. Biointerphases 2015, 10, 040801.
  12. Hirst, A.M.; Frame, F.M.; Arya, M.; Maitland, N.J.; O’Connell, D. Low temperature plasmas as emerging cancer therapeutics: The state of play and thoughts for the future. Tumor Biol. 2016, 37, 7021–7031.
  13. Laroussi, M.; Lu, X.; Keidar, M. Perspective: The Physics, diagnostics, and applications of atmospheric pressure low temperature plasma sources used in plasma medicine. J. Appl. Phys. 2017, 122, 020901.
  14. Kieft, I.E.; Dvinskikh, N.A.; Broers, J.L.v.; Slaaf, D.W.; Stoffels, E. Effect of plasma needle on cultured cells. Proc. SPIE 2004, 5483, 247–251.
  15. Fridman, G.; Shereshevsky, A.; Jost, M.M.; Brooks, A.D.; Fridman, A.; Gutsol, A.; Vasilets, V.; Friedman, G. Floating electrode dielectric barrier discharge plasma in air promoting apoptotic behavior in melanoma skin cancer cell lines. Plasma Chem. Plasma Process. 2007, 27, 163–176.
  16. Lee, H.J.; Shon, C.H.; Kim, Y.S.; Kim, S.; Kim, G.C.; Kong, M.G. Degradation of adhesion molecules of g361 melanoma cells by a non-thermal atmospheric pressure microplasma. New J. Phys. 2009, 11, 115026.
  17. Georgescu, N.; Lupu, A.R. Tumoral and normal cells treatment with high-voltage pulsed cold atmospheric plasma jets. IEEE Trans. Plasma Sci. 2010, 38.8, 1949–1955.
  18. Kim, C.-H.; Bahn, J.H.; Lee, S.-H.; Kim, G.-Y.; Jun, S.-I.; Lee, K.; Baek, S.J. Induction of cell growth arrest by atmospheric non-thermal plasma in colorectal cancer cells. J. Biotechnol. 2010, 150, 530–538.
  19. Kim, G.J.; Kim, W.; Kim, K.T.; Lee, J.K. DNA damage and mitochondria dysfunction in cell apoptosis induced by nonthermal air plasma. Appl. Phys. Lett. 2010, 96, 021502.
  20. Ahn, H.J.; Kim, K.I.; Kim, G.; Moon, E.; Yang, S.S.; Lee, J.S. Atmospheric-pressure plasma jet induces apoptosis involving mitochondria via generation of free radicals. PLoS ONE 2011, 6, e28154.
  21. Köritzer, J.; Boxhammer, V.; Schäfer, A.; Shimizu, T.; Klämpfl, T.G.; Li, Y.F.; Welz, C.; Schwenk-Zieger, S.; Morfill, G.E.; Zimmermann, J.L.; et al. Restoration of sensitivity in chemo-resistant glioma cells by cold atmospheric plasma. PLoS ONE 2013, 8, e64498.
  22. Ja Kim, S.; Min Joh, H.; Chung, T.H. Production of intracellular reactive oxygen species and change of cell viability induced by atmospheric pressure plasma in normal and cancer cells. Appl. Phys. Lett. 2013, 103, 153705.
  23. Arndt, S.; Wacker, E.; Li, Y.F.; Shimizu, T.; Thomas, H.M.; Morfill, G.E.; Karrer, S.; Zimmermann, J.L.; Bosserhoff, A.K. Cold atmospheric plasma, a new strategy to induce senescence in melanoma cells. Exp. Dermatol. 2013, 22, 284–289.
  24. Lin, A.; Truong, B.; Pappas, A.; Kirifides, L.; Oubarri, A.; Chen, S.; Lin, S.; Dobrynin, D.; Fridman, G.; Fridman, A.; et al. Uniform nanosecond pulsed dielectric barrier discharge plasma enhances anti-tumor effects by induction of immunogenic cell death in tumors and stimulation of macrophages. Plasma Processes Polym. 2015, 12, 1392–1399.
  25. Yan, D.; Cui, H.; Zhu, W.; Talbot, A.; Zhang, L.G.; Sherman, J.H.; Keidar, M. The strong cell-based hydrogen peroxide generation triggered by cold atmospheric plasma. Sci. Rep. 2017, 7, 10831.
  26. Shi, L.; Ito, F.; Wang, Y.; Okazaki, Y.; Tanaka, H.; Mizuno, M.; Hori, M.; Hirayama, T.; Nagasawa, H.; Richardson, D.R.; et al. Non-thermal plasma induces a stress response in mesothelioma cells resulting in increased endocytosis, lysosome biogenesis and autophagy. Free Radic. Biol. Med. 2017, 108, 904–917.
  27. Yan, D.; Xu, W.; Yao, X.; Lin, L.; Sherman, J.H.; Keidar, M. The cell activation phenomena in the cold atmospheric plasma cancer treatment. Sci. Rep. 2018, 8, 15418.
  28. Yan, D.; Wang, Q.; Adhikari, M.; Malyavko, A.; Lin, L.; Zolotukhin, D.B.; Yao, X.; Kirschner, M.; Sherman, J.H.; Keidar, M. A physically triggered cell death via transbarrier cold atmospheric plasma cancer treatment. ACS Appl. Mater. Interfaces 2020, 12, 34548–34563.
  29. Yang, X.; Chen, G.; Yu, K.N.; Yang, M.; Peng, S.; Ma, J.; Qin, F.; Cao, W.; Cui, S.; Nie, L.; et al. Cold atmospheric plasma induces GSDME-dependent pyroptotic signaling pathway via ROS generation in tumor cells. Cell Death Dis. 2020, 11, 295.
  30. Yao, X.; Lin, L.; Soni, V.; Gjika, E.; Sherman, J.H.; Yan, D.; Keidar, M. Sensitization of glioblastoma cells to temozolomide by a helium gas discharge tube. Phys. Plasmas 2020, 27, 114502.
  31. Yan, D.; Talbot, A.; Nourmohammadi, N.; Cheng, X.; Canady, J.; Sherman, J.; Keidar, M. Principles of using cold atmospheric plasma stimulated media for cancer treatment. Sci. Rep. 2015, 5, 18339.
  32. Yan, D.; Malyavko, A.; Wang, Q.; Ostrikov, K.K.; Sherman, J.H.; Keidar, M. Multi-modal biological destruction by cold atmospheric plasma: Capability and mechanism. Biomedicines 2021, 9, 1259.
  33. Wang, Q.; Malyavko, A.; Yan, D.; Lamanna, O.K.; Hsieh, M.H.; Sherman, J.; Keidar, M. A Comparative study of cold atmospheric plasma treatment, chemical versus physical strategy. J. Phys. D Appl. Phys. 2020, 54, 095207.
  34. Dezest, M.; Chavatte, L.; Bourdens, M.; Quinton, D.; Camus, M.; Garrigues, L.; Descargues, P.; Arbault, S.; Burlet-Schiltz, O.; Casteilla, L. Mechanistic insights into the impact of cold atmospheric pressure plasma on human epithelial cell lines. Sci. Rep. 2017, 7.1, 41163.
  35. Gorbanev, Y.; O’Connell, D.; Chechik, V. Non-thermal plasma in contact with water: The origin of species. J. Chem. 2016, 22, 3496–3505.
  36. Zhou, R.; Zhou, R.; Wang, P.; Xian, Y.; Mai-prochnow, A. Plasma-activated water: Generation, origin of reactive species and biological applications. J. Phys. D Appl. Phys. 2020, 53, 303001.
  37. Brisset, J.L.; Pawlat, J. Chemical effects of air plasma species on aqueous solutes in direct and delayed exposure modes: Discharge, post-discharge and plasma activated water. Plasma Chem. Plasma Process. 2016, 36, 355–381.
  38. Tanaka, H.; Bekeschus, S.; Yan, D.; Hori, M.; Keidar, M.; Laroussi, M. Plasma-Treated Solutions (PTS) in cancer therapy. Cancers 2021, 13, 1737.
  39. Yan, D.; Horkowitz, A.; Wang, Q.; Keidar, M. On the selective killing of cold atmospheric plasma cancer treatment: Status and beyond. Plasma Processes Polym. 2021, 18, 202100020.
  40. Chernets, N.; Kurpad, D.S.; Alexeev, V.; Rodrigues, D.B.; Freeman, T.A. Reaction chemistry generated by nanosecond pulsed dielectric barrier discharge treatment is responsible for the tumor eradication in the B16 melanoma mouse model. Plasma Processes Polym. 2015, 12, 1400–1409.
  41. Keidar, M.; Walk, R.; Shashurin, A.; Srinivasan, P.; Sandler, A.; Dasgupta, S.; Ravi, R.; Guerrero-Preston, R.; Trink, B. Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy. Br. J. Cancer 2011, 105, 1295–1301.
  42. Binenbaum, Y.; Ben-David, G.; Gil, Z.; Slutsker, Y.Z.; Ryzhkov, M.A.; Felsteiner, J.; Krasik, Y.E.; Cohen, J.T. Cold atmospheric plasma, created at the tip of an elongated flexible capillary using low electric current, can slow the progression of melanoma. PLoS ONE 2017, 12, e0169457.
  43. Boehm, D.; Heslin, C.; Cullen, P.J.; Bourke, P. Cytotoxic and mutagenic potential of solutions exposed to cold atmospheric plasma. Sci. Rep. 2016, 6, 21464.
  44. Yan, D.; Malyavko, A.; Wang, Q.; Lin, L.; Sherman, J.H. Applied sciences cold atmospheric plasma cancer treatment, a critical review. Appl. Sci. 2021, 11, 7757.
  45. Vandamme, M.; Robert, E.; Pesnel, S.; Barbosa, E.; Dozias, S.; Sobilo, J.; Lerondel, S.; le Pape, A.; Pouvesle, J.M. Antitumor effect of plasma treatment on U87 glioma xenografts: Preliminary results. Plasma Processes Polym. 2010, 7, 264–273.
  46. Vandamme, M.; Robert, E.; Dozias, S.; Sobilo, J.; Lerondel, S.; le Pape, A.; Pouvesle, J.-M. Response of human glioma U87 xenografted on mice to non thermal plasma treatment. Plasma Med. 2011, 1, 27–43.
  47. Brullé, L.; Vandamme, M.; Riès, D.; Martel, E.; Robert, E.; Lerondel, S.; Trichet, V.; Richard, S.; Pouvesle, J.M.; le Pape, A. Effects of a non thermal plasma treatment alone or in combination with gemcitabine in a MIA PaCa2-luc orthotopic pancreatic carcinoma model. PLoS ONE 2012, 7, e52653.
  48. Vandamme, M.; Robert, E.; Lerondel, S.; Sarron, V.; Ries, D.; Dozias, S.; Sobilo, J.; Gosset, D.; Kieda, C.; Legrain, B.; et al. ROS implication in a new antitumor strategy based on non-thermal plasma. Int. J. Cancer. 2012, 130, 2185–2194.
  49. Walk, R.M.; Snyder, J.A.; Srinivasan, P.; Kirsch, J.; Diaz, S.O.; Blanco, F.C.; Shashurin, A.; Keidar, M.; Sandler, A.D. Cold atmospheric plasma for the ablative treatment of neuroblastoma. J. Pediatr. Surg. 2013, 48, 67–73.
  50. Yajima, I.; Iida, M.; Kumasaka, M.Y.; Omata, Y.; Ohgami, N.; Chang, J.; Ichihara, S.; Hori, M.; Kato, M. Non-equilibrium atmospheric pressure plasmas modulate cell cycle-related gene expressions in melanocytic tumors of RET-transgenic mice. Exp. Dermatol. 2014, 23, 424–425.
  51. Kang, S.U.; Cho, J.-H.; Chang, J.W.; Shin, Y.S.; Kim, K.I.; Park, J.K.; Yang, S.S.; Lee, J.-S.; Moon, E.; Lee, K.; et al. Nonthermal plasma induces head and neck cancer cell death: The potential involvement of mitogen-activated protein kinase-dependent mitochondrial reactive oxygen species. Cell Death Dis. 2014, 5, e1056.
  52. Ikeda, J.I.; Tsuruta, Y.; Nojima, S.; Sakakita, H.; Hori, M.; Ikehara, Y. Anti-cancer effects of nonequilibrium atmospheric pressure plasma on cancer-initiating cells in human endometrioid adenocarcinoma cells. Plasma Processes Polym. 2015, 12, 1370–1376.
  53. Kaushik, N.K.; Kaushik, N.; Yoo, K.C.; Uddin, N.; Kim, J.S.; Lee, S.J.; Choi, E.H. Low doses of PEG-coated gold nanoparticles sensitize solid tumors to cold plasma by blocking the PI3K/AKT-driven signaling axis to suppress cellular transformation by inhibiting growth and EMT. Biomaterials 2016, 87, 118–130.
  54. Mirpour, S.; Piroozmand, S.; Soleimani, N.; Jalali Faharani, N.; Ghomi, H.; Fotovat Eskandari, H.; Sharifi, A.M.; Mirpour, S.; Eftekhari, M.; Nikkhah, M. Utilizing the micron sized non-thermal atmospheric pressure plasma inside the animal body for the tumor treatment application. Sci. Rep. 2016, 6, 29048.
  55. Lin, A.G.; Xiang, B.; Merlino, D.J.; Baybutt, T.R.; Sahu, J.; Fridman, A.; Snook, A.E.; Miller, V. Non-thermal plasma induces immunogenic cell death in vivo in murine CT26 colorectal tumors. Oncoimmunology 2018, 7, e1484978.
More
ScholarVision Creations