Skip to main content

Advertisement

Log in

Intraperitoneal Administration of Plasma-Activated Medium: Proposal of a Novel Treatment Option for Peritoneal Metastasis From Gastric Cancer

  • Gastrointestinal Oncology
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background

The administration of fluid irradiated with non-equilibrium atmospheric pressure plasma (NEAPP) has attracted much interest as a novel therapeutic method for cancer. The authors previously reported on the efficacy of plasma-activated medium (PAM) for treating cancer cell lines through the induction of apoptosis. In this study, the therapeutic effect of PAM was evaluated in vivo using a peritoneal metastasis mouse model.

Methods

Two gastric cancer cell lines were used in proliferation assays performed to optimize the production of PAM by changing the distance between the plasma source and the medium surface and by altering the volume of irradiated medium. Wound-healing and adhesion assays were conducted to determine the effect of PAM therapy on cell migration and adhesion capacity in vitro. Finally, a mouse model established by the intraperitoneal injection of enhanced green fluorescent protein-tagged gastric cancer cells was used to explore the efficacy of PAM administered intraperitoneally in inhibiting peritoneal metastasis formation.

Results

Shorter distances between the plasma source and the medium surface and smaller volumes of treated medium increased the anti-tumor effect of PAM. The PAM treatment attenuated gastric cancer cell migration and adhesion in vitro. The intraperitoneal administration of PAM decreased the formation of peritoneal metastatic nodules by 60% in the mouse model, and no adverse events were observed.

Conclusions

Plasma-activated liquids may represent a novel therapeutic method for the treatment of peritoneal metastases in gastric cancer.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Jung KW, Won YJ, Kong HJ, Oh CM, Lee DH, Lee JS. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2011. Cancer Res Treat. 2014;46:109–23.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9–29.

    Article  PubMed  Google Scholar 

  3. Shiozaki H, Elimova E, Slack RS, et al. Prognosis of gastric adenocarcinoma patients with various burdens of peritoneal metastases. J Surg Oncol. 2015;113:29–35.

    Article  PubMed  Google Scholar 

  4. Brun P, Brun P, Vono M, et al. Disinfection of ocular cells and tissues by atmospheric-pressure cold plasma. PLoS One. 2012;7:e33245.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Fridman G, Friedman G, Gutsol A, Shekhter AB, Vasilets VN, Fridman A. Applied plasma medicine. Plasma Process Polym. 2008;5:503–33.

    Article  CAS  Google Scholar 

  6. Fridman G, Peddinghaus M, Balasubramanian M, Ayan H, Fridman A, Gutsol A, et al. Blood coagulation and living tissue sterilization by floating-electrode dielectric barrier discharge in air. Plasma Chem Plasma Proc. 2006;26:425–42.

    Article  CAS  Google Scholar 

  7. Ahn HJ, Kim KI, Kim G, Moon E, Yang SS, Lee JS. Atmospheric-pressure plasma jet induces apoptosis involving mitochondria via generation of free radicals. PLoS One. 2011;6:e28154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gesbert F, Larue L. Non-thermal plasmas: novel preventive and curative therapy against melanomas? Exp Dermatol. 2014;23:716–7.

    Article  PubMed  Google Scholar 

  9. Hou J, Ma J, Yu KN, Li W, Cheng C, Bao L, Han W. Nonthermal plasma treatment altered gene expression profiling in non-small cell lung cancer A549 cells. BMC Genomics. 2015;16:435.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ma Y, Ha CS, Hwang SW, Lee HJ, Kim GC, Lee KW, Song K. Nonthermal atmospheric pressure plasma preferentially induces apoptosis in p53-mutated cancer cells by activating ROS stress-response pathways. PLoS One. 2014;9:e91947.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Thiyagarajan M, Sarani A, Gonzales XF. Characterization of an atmospheric pressure plasma jet and its applications for disinfection and cancer treatment. Stud Health Technol Inform. 2013;184:443–9.

    PubMed  Google Scholar 

  12. Tanaka H, Mizuno M, Ishikawa K, et al. Plasma with high electron density and plasma-activated medium for cancer treatment. Clin Plasma Med. 2015;3:72–6.

    Article  Google Scholar 

  13. Tanaka H, Mizuno M, Toyokuni S, et al. Cancer therapy using nonthermal atmospheric pressure plasma with ultra-high electron density. Phys Plasmas. 2015;22:122004.

    Article  Google Scholar 

  14. Tanaka H, Mizuno M, Ishikawa K, et al. Plasma-activated medium selectively kills glioblastoma brain tumor cells by downregulating a survival signaling molecule, AKT kinase. Plasma Med. 2011;1:265–77.

    Article  Google Scholar 

  15. Hattori N, Yamada S, Torii K, et al. Effectiveness of plasma treatment on pancreatic cancer cells. Int J Oncol. 2015;47:1655–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Torii K, Yamada S, Nakamura K, et al. Effectiveness of plasma treatment on gastric cancer cells. Gastric Cancer. 2014;18:635–43.

    Article  PubMed  Google Scholar 

  17. Utsumi F, Kajiyama H, Nakamura K, Tanaka H, Hori M, Kikkawa F. Selective cytotoxicity of indirect nonequilibrium atmospheric pressure plasma against ovarian clear-cell carcinoma. Springerplus. 2014;3:398.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Chang JW, Kang SU, Shin YS, et al. Non-thermal atmospheric pressure plasma inhibits thyroid papillary cancer cell invasion via cytoskeletal modulation, altered MMP-2/-9/uPA activity. PLoS One. 2014;9:e92198.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Kim CH, Kwon S, Bahn JH, Lee K, Jun SI, Rack PD, Baek SJ. Effects of atmospheric nonthermal plasma on invasion of colorectal cancer cells. Appl Phys Lett. 2010;96:243701.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Utsumi F, Kajiyama H, Nakamura K, et al. Effect of indirect nonequilibrium atmospheric pressure plasma on anti-proliferative activity against chronic chemo-resistant ovarian cancer cells in vitro and in vivo. PLoS One. 2013;8:e81576.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Ohashi N, Kodera Y, Nakanishi H, et al. Efficacy of intraperitoneal chemotherapy with paclitaxel targeting peritoneal micrometastasis as revealed by GFP-tagged human gastric cancer cell lines in nude mice. Int J Oncol. 2005;27:637–44.

    CAS  PubMed  Google Scholar 

  22. Kieft IE, Broers JLV, Caubet-Hilloutou V, Slaaf DW, Ramaekers FCS, Stoffels E. Electric discharge plasmas influence attachment of cultured CHO K1 cells. Bioelectromagnetics. 2004;25:362–8.

    Article  CAS  PubMed  Google Scholar 

  23. Keidar M, Walk R, Shashurin A, et al. Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy. Br J Cancer. 2011;105:1295–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Vandamme M, Robert E, Dozias S, Sobilo J, Lerondel S, Le Pape A, et al. Response of human glioma U87 xenografted on mice to nonthermal plasma treatment. Plasma Med. 2011;1:27–43.

    Article  Google Scholar 

  25. Brulle L, Vandamme M, Ries D, et al. Effects of a nonthermal plasma treatment alone or in combination with gemcitabine in a MIA PaCa2-luc orthotopic pancreatic carcinoma model. PLoS One. 2012;7:e52653.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Vandamme M, Robert E, Lerondel S, et al. ROS implication in a new antitumor strategy based on nonthermal plasma. Int J Cancer. 2012;130:2185–94.

    Article  CAS  PubMed  Google Scholar 

  27. Walk RM, Snyder JA, Srinivasan P, et al. Cold atmospheric plasma for the ablative treatment of neuroblastoma. J Pediatr Surg. 2013;48:67–73.

    Article  PubMed  Google Scholar 

  28. Utsumi F, Kajiyama H, Nakamura KAE, et al. Variable susceptibility of ovarian cancer cells to nonthermal plasma-activated medium. Oncol Rep. 2016;35:3169–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Joh HM, Choi JY, Kim SJ, Chung TH, Kang TH. Effect of additive oxygen gas on cellular response of lung cancer cells induced by atmospheric-pressure helium plasma jet. Sci Rep. 2014;4:6638.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Winter J, Wende K, Masur K, et al. Feed gas humidity: a vital parameter affecting a cold atmospheric-pressure plasma jet and plasma-treated human skin cells. J Phys D Appl Phys. 2013;46:295401.

    Article  Google Scholar 

  31. Rice BW, Cable MD, Nelson MB. In vivo imaging of light-emitting probes. J Biomed Opt. 2001;6:432–40.

    Article  CAS  PubMed  Google Scholar 

  32. Kishimoto H, Zhao M, Hayashi K, et al. In vivo internal tumor illumination by telomerase-dependent adenoviral GFP for precise surgical navigation. Proc Natl Acad Sci USA. 2009;106:14514–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kishimoto H, Aki R, Urata Y, et al. Tumor-selective, adenoviral-mediated GFP genetic labeling of human cancer in the live mouse reports future recurrence after resection. Cell Cycle. 2011;10:2737–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Weissleder R. A clearer vision for in vivo imaging. Nat Biotechnol. 2001;19:316–7.

    Article  CAS  PubMed  Google Scholar 

Download references

Disclosures

There are no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Suguru Yamada MD, PhD, FACS.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10434_2016_5759_MOESM1_ESM.ppt

Supplementary material 1 (PPT 845 kb) Experimental system for producing PAM. (a) Photograph of our device. (b) Schematics of PAM production.

10434_2016_5759_MOESM2_ESM.pptx

Supplementary material 2 (PPTX 73 kb)Effect of repeated PAM exposure. A total of 3×103 cells were seeded into a 96-well plate, and the medium was replaced as follows. Untreated: The medium was replaced every 24 h with RPMI-1640. 1: At 24 h after seeding, the medium was replaced with PAM, and after another 24 h and 48 h, the medium was replaced with RPMI-1640. 2: At 24 h and 48 h after seeding, the medium was replaced with PAM, and after another 24 h, the medium was replaced with RPMI-1640. 3: At 24 h, 48 h and 72 h after seeding, the medium was replaced with PAM. Finally, at 96 h after seeding, an MTS assay was performed, and the proliferation rates were calculated. Each column represents the mean of three replicates, and the error bars represent standard deviation.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Takeda, S., Yamada, S., Hattori, N. et al. Intraperitoneal Administration of Plasma-Activated Medium: Proposal of a Novel Treatment Option for Peritoneal Metastasis From Gastric Cancer. Ann Surg Oncol 24, 1188–1194 (2017). https://doi.org/10.1245/s10434-016-5759-1

Download citation

  • Received:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-016-5759-1

Keywords

Navigation