Cold Atmospheric Plasma: A New Strategy Based Primarily on Oxidative Stress for Osteosarcoma Therapy
Abstract
:1. Background
2. Current Treatment in OS
2.1. Surgery
2.2. Chemotherapy
2.3. Radiotherapy
2.4. Other Therapeutic Options
3. CAP for Cancer Therapy
3.1. Application Methods of Cold Atmospheric Plasma (CAP)
3.2. Anti-Cancer Mechanism of CAP
3.3. Advantages of Using CAP
4. Potential Application of CAP in OS
5. In Vitro Effects of CAP in OS
5.1. Direct Treatment in OS
5.2. Indirect Treatment in OS
6. Challenges of CAP for OS Therapy
6.1. Bone Microenvironment in OS
6.2. Tumor Heterogeneity in OS
6.2.1. Oncogenes in OS
6.2.2. How Could CAP Affect Intracellular Signaling in OS?
6.2.3. Cancer Stem Cells in OS
6.2.4. How Could CAP-Induced Oxidative Stress Affect CSCs in OS?
7. Future Trends
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
ALDH | Aldehyde dehydrogenase |
CAP | Cold atmospheric plasma |
CAF | Cancer associated fibroblast |
CSC | Cancer stem cell |
ECM | Extra-cellular matrix |
HSP | Heat-shock protein |
IL | Interleukin |
MSC | Mesenchymal stem cell |
OS | Osteosarcoma |
pRB | Retinoblastoma |
RNS | Reactive nitrogen species |
RONS | Reactive oxygen and nitrogen species |
ROS | Reactive oxygen species |
UV | Ultraviolet |
VEGF | Vascular endothelial growth factor |
References
- Gill, J.; Ahluwalia, M.K.; Geller, D.; Gorlick, R. New targets and approaches in osteosarcoma. Pharmacol. Ther. 2013, 137, 89–99. [Google Scholar] [CrossRef]
- Parry, M.C.; Laitinen, M.; Albergo, J.; Jeys, L.; Carter, S.; Gaston, C.L.; Sumathi, V.; Grimer, R.J. Osteosarcoma of the pelvis. Bone Joint J. 2016, 98, 555–563. [Google Scholar] [CrossRef]
- Yao, Z.; Han, L.; Chen, Y.; He, F.; Sun, B.; Kamar, S.; Zhang, Y.; Yang, Y.; Wang, C.; Yang, Z. Hedgehog signalling in the tumourigenesis and metastasis of osteosarcoma, and its potential value in the clinical therapy of osteosarcoma review. Cell Death Dis. 2018, 9, 1–12. [Google Scholar]
- Meazza, C.; Scanagatta, P. Metastatic osteosarcoma: A challenging multidisciplinary treatment. Expert Rev. Anticancer Ther. 2016, 16, 543–556. [Google Scholar] [CrossRef]
- Spraker-Perlman, H.L.; Barkauskas, D.A.; Krailo, M.D.; Meyers, P.A.; Schwartz, C.L.; Doski, J.; Gorlick, R.; Janeway, K.A.; Isakoff, M.S. Factors influencing survival after recurrence in osteosarcoma: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2019, 66, e27444. [Google Scholar] [CrossRef]
- Allison, D.C.; Carney, S.C.; Ahlmann, E.R.; Hendifar, A.; Chawla, S.; Fedenko, A.; Angeles, C.; Menendez, L.R. A Meta-Analysis of Osteosarcoma Outcomes in the Modern Medical Era. Sarcoma 2012, 2012. [Google Scholar] [CrossRef] [Green Version]
- Botter, S.M.; Neri, D.; Fuchs, B. Recent advances in osteosarcoma. Curr. Opin. Pharmacol. 2014, 16, 15–23. [Google Scholar] [CrossRef]
- Isakoff, M.S.; Bielack, S.S.; Meltzer, P.; Gorlick, R. Osteosarcoma: Current Treatment and a Collaborative Pathway to Success. J. Clin. Oncol. 2015, 33, 3029–3035. [Google Scholar] [CrossRef] [Green Version]
- Dubuc, A.; Monsarrat, P.; Virard, F.; Merbahi, N.; Sarrette, J.-P.; Laurencin-Dalicieux, S.; Cousty, S. Use of cold-atmospheric plasma in oncology: A concise systematic review. Ther. Adv. Med. Oncol. 2018, 10. [Google Scholar] [CrossRef]
- Schuster, M.; Seebauer, C.; Rutkowski, R.; Hauschild, A.; Podmelle, F.; Metelmann, C.; Metelmann, B.; von Woedtke, T.; Hasse, S.; Weltmann, K.-D.; et al. Visible tumor surface response to physical plasma and apoptotic cell kill in head and neck cancer. J. Cranio-Maxillofacial Surg. 2016, 44, 1445–1452. [Google Scholar] [CrossRef]
- Anderson, M.E. Update on Survival in Osteosarcoma. Orthop. Clin. North Am. 2016, 47, 283–292. [Google Scholar]
- Durfee, R.A.; Mohammed, M.; Luu, H.H. Review of Osteosarcoma and Current Management. Rheumatol. Ther. 2016, 3, 221–243. [Google Scholar] [CrossRef] [Green Version]
- Panagopoulos, G.N.; Mavrogenis, A.F.; Mauffrey, C.; Lesenský, J.; Angelini, A.; Megaloikonomos, P.D.; Igoumenou, V.G.; Papanastassiou, J.; Savvidou, O.; Ruggieri, P.; et al. Intercalary reconstructions after bone tumor resections: A review of treatments. Eur. J. Orthop. Surg. Traumatol. 2017, 27, 737–746. [Google Scholar]
- Zhang, Y.; Yang, J.; Zhao, N.; Wang, C.; Kamar, S.; Zhou, Y.; He, Z.; Yang, J.; Sun, B.; Shi, X.; et al. Progress in the chemotherapeutic treatment of osteosarcoma (Review). Oncol. Lett. 2018, 16, 6228–6237. [Google Scholar] [CrossRef] [Green Version]
- Lopes-Júnior, L.C.; Sayuri, D.; Vulczak, A.; Cristina, J. Emerging Cytokine Networks in Osteosarcoma. Cancer Cell Microenviron. 2017. [Google Scholar] [CrossRef] [Green Version]
- Howard, S.C.; McCormick, J.; Pui, C.; Buddington, R.K.; Harvey, R.D. Preventing and Managing Toxicities of High-Dose Methotrexate. Oncologist 2016, 21, 1471–1482. [Google Scholar] [CrossRef] [Green Version]
- Rivankar, S. An overview of doxorubicin formulations in cancer therapy. J. Cancer Res. Ther. 2014, 10, 853. [Google Scholar] [CrossRef]
- Dasari, S.; Bernard Tchounwou, P. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar]
- Armstrong, J.; Dass, C.R. Doxorubicin Action on Mitochondria: Relevance to Osteosarcoma Therapy? Curr. Drug Targets 2018, 19, 432–438. [Google Scholar] [CrossRef]
- Taneja, N.; Tjalkens, R.; Philbert, M.A.; Rehemtulla, A. Irradiation of mitochondria initiates apoptosis in cell free system. Oncogene 2001, 20, 167–177. [Google Scholar] [CrossRef] [Green Version]
- Oertel, S.; Blattmann, C.; Rieken, S.; Jensen, A.; Combs, S.E.; Huber, P.E.; Bischof, M.; Kulozik, A.; Debus, J.; Schulz-Ertner, D. Radiotherapy in the treatment of primary osteosarcoma—A single center experience. Tumori 2010, 96, 582–588. [Google Scholar] [CrossRef]
- DeLaney, T.F.; Park, L.; Goldberg, S.I.; Hug, E.B.; Liebsch, N.J.; Munzenrider, J.E.; Suit, H.D. Radiotherapy for local control of osteosarcoma. Int. J. Radiat. Oncol. Biol. Phys. 2005, 61, 492–498. [Google Scholar] [CrossRef]
- Habash, M.; Bohorquez, L.C.; Kyriakou, E.; Kron, T.; Martin, O.A.; Blyth, B.J. Clinical and functional assays of radiosensitivity and radiation-induced second cancer. Cancers 2017, 9, 147. [Google Scholar] [CrossRef] [Green Version]
- Echchikhi, Y.; Loughlimi, H.; Touil, A.; Kebdani, T.; Benjaafar, N. Radiation-induced osteosarcoma of the skull base after radiation therapy in a patient with nasopharyngeal carcinoma: A case report and review of the literature. J. Med. Case Rep. 2016, 10, 334. [Google Scholar] [CrossRef] [Green Version]
- Futamura, G.; Kawabata, S.; Siba, H.; Kuroiwa, T.; Suzuki, M.; Kondo, N.; Ono, K.; Sakurai, Y.; Tanaka, M.; Todo, T.; et al. A case of radiation-induced osteosarcoma treated effectively by boron neutron capture therapy. Radiat. Oncol. 2014, 9, 237. [Google Scholar] [CrossRef] [Green Version]
- Rettew, A.N.; Getty, P.J.; Greenfield, E.M. Receptor Tyrosine Kinases in Osteosarcoma: Not Just the Usual Suspects; Springer: Cham, Switzerland, 2014; pp. 47–66. [Google Scholar]
- Zhang, S.; Wang, X.; Gu, Z.; Wang, L. Small Molecule Survivin Inhibitor YM155 Displays Potent Activity Against Human Osteosarcoma Cells. Cancer Invest. 2016, 34, 401–407. [Google Scholar] [CrossRef]
- Onimoe, G.-I.; Liu, A.; Lin, L.; Wei, C.-C.; Schwartz, E.B.; Bhasin, D.; Li, C.; Fuchs, J.R.; Li, P.; Houghton, P.; et al. Small molecules, LLL12 and FLLL32, inhibit STAT3 and exhibit potent growth suppressive activity in osteosarcoma cells and tumor growth in mice. Invest. New Drugs 2012, 30, 916–926. [Google Scholar] [CrossRef]
- Jian, F.; Yuan, F.; Jiong, M.; Zhu, X.; Yu, G.; Lu, D. Silencing of Glucose Transporter Protein-1 by RNA Interference Inhibits Human Osteosarcoma Mg63 Cells Growth in vivo. Technol. Cancer Res. Treat. 2015, 14, 243–248. [Google Scholar] [CrossRef]
- Xie, X.; Ye, Z.; Yang, D.; Tao, H. Effects of combined c-myc and Bmi-1 siRNAs on the growth and chemosensitivity of MG-63 osteosarcoma cells. Mol. Med. Rep. 2013, 8, 168–172. [Google Scholar] [CrossRef]
- Wedekind, M.F.; Wagner, L.M.; Cripe, T.P. Immunotherapy for osteosarcoma: Where do we go from here? Pediatr. Blood Cancer 2018, 65, e27227. [Google Scholar] [CrossRef] [Green Version]
- Varshney, J.; Scott, M.C.; Largaespada, D.A.; Subramanian, S. Understanding the osteosarcoma pathobiology: A comparative oncology approach. Vet. Sci. 2016, 3, 3. [Google Scholar] [CrossRef]
- Bishop, M.W.; Janeway, K.A.; Gorlick, R. Future directions in the treatment of osteosarcoma. Curr. Opin. Pediatr. 2016, 28, 26–33. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, C.; Berganza, C.; Zhang, J. Cold Atmospheric Plasma: Methods of production and application in dentistry and oncology. Med. Gas Res. 2013, 3, 21. [Google Scholar] [CrossRef] [Green Version]
- Setsuhara, Y. Low-temperature atmospheric-pressure plasma sources for plasma medicine. Arch. Biochem. Biophys. 2016, 605, 3–10. [Google Scholar] [CrossRef]
- Mashayekh, S.; Rajaee, H.; Akhlaghi, M.; Shokri, B.; Hassan, Z.M. Atmospheric-pressure plasma jet characterization and applications on melanoma cancer treatment (B/16-F10). Phys. Plasmas 2015, 22, 093508. [Google Scholar] [CrossRef]
- Lackmann, J.-W.; Bandow, J.E. Inactivation of microbes and macromolecules by atmospheric-pressure plasma jets. Appl. Microbiol. Biotechnol. 2014, 98, 6205–6213. [Google Scholar] [CrossRef]
- Wiegand, C.; Beier, O.; Horn, K.; Pfuch, A.; Tölke, T.; Hipler, U.-C.; Schimanski, A. Antimicrobial Impact of Cold Atmospheric Pressure Plasma on Medical Critical Yeasts and Bacteria Cultures. Skin Pharmacol. Physiol. 2014, 27, 25–35. [Google Scholar] [CrossRef]
- Xu, G.-M.; Shi, X.-M.; Cai, J.-F.; Chen, S.-L.; Li, P.; Yao, C.-W.; Chang, Z.-S.; Zhang, G.-J. Dual effects of atmospheric pressure plasma jet on skin wound healing of mice. Wound Repair Regen. 2015, 23, 878–884. [Google Scholar] [CrossRef]
- Arndt, S.; Unger, P.; Wacker, E.; Shimizu, T.; Heinlin, J.; Li, Y.-F.; Thomas, H.M.; Morfill, G.E.; Zimmermann, J.L.; Bosserhoff, A.-K.; et al. Cold Atmospheric Plasma (CAP) Changes Gene Expression of Key Molecules of the Wound Healing Machinery and Improves Wound Healing In Vitro and In Vivo. PLoS ONE 2013, 8, e79325. [Google Scholar] [CrossRef] [Green Version]
- Steuer, A.; Wolff, C.M.; Von Woedtke, T.; Weltmann, K.D.; Kolb, J.F. Cell stimulation versus cell death induced by sequential treatments with pulsed electric fields and cold atmospheric pressure plasma. PLoS ONE 2018, 13. [Google Scholar] [CrossRef]
- Chen, Z.; Simonyan, H.; Cheng, X.; Gjika, E.; Lin, L.; Canady, J.; Sherman, J.H.; Young, C.; Keidar, M. A Novel Micro Cold Atmospheric Plasma Device for Glioblastoma Both In Vitro and In Vivo. Cancers 2017, 9, 61. [Google Scholar] [CrossRef] [Green Version]
- Xiang, L.; Xu, X.; Zhang, S.; Cai, D.; Dai, X. Cold atmospheric plasma conveys selectivity on triple negative breast cancer cells both in vitro and in vivo. Free Radic. Biol. Med. 2018, 124, 205–213. [Google Scholar] [CrossRef]
- Metelmann, H.R.; Seebauer, C.; Miller, V.; Fridman, A.; Bauer, G.; Graves, D.B.; Pouvesle, J.M.; Rutkowski, R.; Schuster, M.; Bekeschus, S.; et al. Clinical experience with cold plasma in the treatment of locally advanced head and neck cancer. Clin. Plasma Med. 2018, 9, 6–13. [Google Scholar] [CrossRef]
- Zou, P.; Zhang, J.; Xia, Y.; Kanchana, K.; Guo, G.; Chen, W.; Huang, Y.; Wang, Z.; Yang, S.; Liang, G. ROS generation mediates the anti-cancer effects of WZ35 via activating JNK and ER stress apoptotic pathways in gastric cancer. Oncotarget 2015, 6, 5860–5876. [Google Scholar] [CrossRef] [Green Version]
- Akter, M.; Jangra, A.; Choi, S.A.; Choi, E.H.; Han, I. Non-thermal atmospheric pressure bio-compatible plasma stimulates apoptosis via p38/MAPK mechanism in U87 malignant glioblastoma. Cancers 2020, 12, 245. [Google Scholar] [CrossRef] [Green Version]
- Kaushik, N.; Uddin, N.; Sim, G.B.; Hong, Y.J.; Baik, K.Y.; Kim, C.H.; Lee, S.J.; Kaushik, N.K.; Choi, E.H. Responses of solid tumor cells in DMEM to reactive oxygen species generated by non-thermal plasma and chemically induced ROS systems. Sci. Rep. 2015, 5, 8587. [Google Scholar] [CrossRef] [Green Version]
- Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2017. [Google Scholar] [CrossRef]
- Attri, P.; Park, J.H.; Ali, A.; Choi, E.H. How Does Plasma Activated Media Treatment Differ From Direct Cold Plasma Treatment? Anticancer. Agents Med. Chem. 2018, 18, 805–814. [Google Scholar] [CrossRef]
- Yusupov, M.; Van der Paal, J.; Neyts, E.C.; Bogaerts, A. Synergistic effect of electric field and lipid oxidation on the permeability of cell membranes. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 839–847. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Privat-Maldonado, A.; Gorbanev, Y.; Dewilde, S.; Smits, E.; Bogaerts, A. Reduction of Human Glioblastoma Spheroids Using Cold Atmospheric Plasma: The Combined Effect of Short- and Long-Lived Reactive Species. Cancers 2018, 10, 394. [Google Scholar] [CrossRef] [Green Version]
- Saadati, F.; Mahdikia, H.; Abbaszadeh, H.-A.; Abdollahifar, M.-A.; Khoramgah, M.S.; Shokri, B. Comparison of Direct and Indirect cold atmospheric-pressure plasma methods in the B16F10 melanoma cancer cells treatment. Sci. Rep. 2018, 8, 7689. [Google Scholar] [CrossRef] [Green Version]
- Partecke, L.I.; Evert, K.; Haugk, J.; Doering, F.; Normann, L.; Diedrich, S.; Weiss, F.U.; Evert, M.; Huebner, N.O.; Guenther, C.; et al. Tissue Tolerable Plasma (TTP) induces apoptosis in pancreatic cancer cells in vitro and in vivo. BMC Cancer 2012, 12, 473. [Google Scholar] [CrossRef] [Green Version]
- Graves, D.B. Reactive Species from Cold Atmospheric Plasma: Implications for Cancer Therapy. Plasma Process. Polym. 2014, 11, 1120–1127. [Google Scholar] [CrossRef]
- Khlyustova, A.; Labay, C.; Machala, Z.; Ginebra, M.-P.; Canal, C. Important parameters in plasma jets for the production of RONS in liquids for plasma medicine: A brief review. Front. Chem. Sci. Eng. 2019. [Google Scholar] [CrossRef]
- Chauvin, J.; Judée, F.; Yousfi, M.; Vicendo, P.; Merbahi, N. Analysis of reactive oxygen and nitrogen species generated in three liquid media by low temperature helium plasma jet. Sci. Rep. 2017, 7, 4562. [Google Scholar] [CrossRef]
- Tanaka, H.; Mizuno, M.; Kikkawa, F.; Hori, M. Plasma-activated medium and its medical and biological applications. In Proceedings of the 2016 IEEE International Conference on Plasma Science (ICOPS), Banff, AB, Canada, 19–23 June 2016; p. 1. [Google Scholar]
- Jawaid, P.; Rehman, M.U.; Zhao, Q.L.; Takeda, K.; Ishikawa, K.; Hori, M.; Shimizu, T.; Kondo, T. Helium-based cold atmospheric plasma-induced reactive oxygen species-mediated apoptotic pathway attenuated by platinum nanoparticles. J. Cell. Mol. Med. 2016, 20, 1737–1748. [Google Scholar] [CrossRef]
- Volotskova, O.; Hawley, T.S.; Stepp, M.A.; Keidar, M. Targeting the cancer cell cycle by cold atmospheric plasma. Sci. Rep. 2012, 2, 636. [Google Scholar] [CrossRef] [Green Version]
- Aryal, S.; Bisht, G. New Paradigm for a Targeted Cancer Therapeutic Approach: A Short Review on Potential Synergy of Gold Nanoparticles and Cold Atmospheric Plasma. Biomedicines 2017, 5, 38. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- Kamm, A.; Przychodzen, P.; Kuban-Jankowska, A.; Jacewicz, D.; Dabrowska, A.M.; Nussberger, S.; Wozniak, M.; Gorska-Ponikowska, M. Nitric oxide and its derivatives in the cancer battlefield. Nitric Oxide Biol. Chem. 2019, 93, 102–114. [Google Scholar]
- Ahn, H.J.; Kim, K.; 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. [Google Scholar] [CrossRef] [Green Version]
- Iuchi, K.; Morisada, Y.; Yoshino, Y.; Himuro, T.; Saito, Y.; Murakami, T.; Hisatomi, H. Cold atmospheric-pressure nitrogen plasma induces the production of reactive nitrogen species and cell death by increasing intracellular calcium in HEK293T cells. Arch. Biochem. Biophys. 2018, 654, 136–145. [Google Scholar] [CrossRef]
- Privat-Maldonado, A.; Schmidt, A.; Lin, A.; Weltmann, K.-D.; Wende, K.; Bogaerts, A.; Bekeschus, S. ROS from Physical Plasmas: Redox Chemistry for Biomedical Therapy. Oxid. Med. Cell. Longev. 2019, 2019. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Panngom, K.; Baik, K.; Nam, M.; Han, J.; Rhim, H.; Choi, E. Preferential killing of human lung cancer cell lines with mitochondrial dysfunction by nonthermal dielectric barrier discharge plasma. Cell Death Dis. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
- Choi, J.Y.; Joh, H.M.; Park, J.-M.; Kim, M.J.; Chung, T.H.; Kang, T.-H. Non-thermal plasma-induced apoptosis is modulated by ATR- and PARP1-mediated DNA damage responses and circadian clock. Oncotarget 2016, 7, 32980–32989. [Google Scholar] [CrossRef]
- Schneider, C.; Gebhardt, L.; Arndt, S.; Karrer, S.; Zimmermann, J.L.; Fischer, M.J.M.; Bosserhoff, A.-K. Cold atmospheric plasma causes a calcium influx in melanoma cells triggering CAP-induced senescence. Sci. Rep. 2018, 8, 10048. [Google Scholar] [CrossRef]
- Zhou, R.; Zhou, R.; Zhuang, J.; Zong, Z.; Zhang, X.; Liu, D.; Bazaka, K.; Ostrikov, K. Interaction of Atmospheric-Pressure Air Microplasmas with Amino Acids as Fundamental Processes in Aqueous Solution. PLoS ONE 2016, 11, e0155584. [Google Scholar] [CrossRef] [Green Version]
- Bauer, G.; Sersenová, D.; Graves, D.B.; Machala, Z. Cold Atmospheric Plasma and Plasma-Activated Medium Trigger RONS-Based Tumor Cell Apoptosis. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef]
- Conway, G.E.; Casey, A.; Milosavljevic, V.; Liu, Y.; Howe, O.; Cullen, P.J.; Curtin, J.F. Non-thermal atmospheric plasma induces ROS-independent cell death in U373MG glioma cells and augments the cytotoxicity of temozolomide. Br. J. Cancer 2016, 114, 435–443. [Google Scholar] [CrossRef]
- Schneider, C.; Arndta, S.; Zimmermann, J.L.; Li, Y.; Karrer, S.; Bosserhoff, A.K. Cold atmospheric plasma treatment inhibits growth in colorectal cancer cells. Biol. Chem. 2018, 400, 111–127. [Google Scholar] [CrossRef]
- Dezest, M.; Chavatte, L.; Bourdens, M.; Quinton, D.; Camus, M.; Garrigues, L.; Descargues, P.; Arbault, S.; Burlet-Schiltz, O.; Casteilla, L.; et al. Mechanistic insights into the impact of Cold Atmospheric Pressure Plasma on human epithelial cell lines. Sci. Rep. 2017, 7, 41163. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- Ahn, H.J.; Kim, K.I.; Hoan, N.N.; Kim, C.H.; Moon, E.; Choi, K.S.; Yang, S.S.; Lee, J.S. Targeting cancer cells with reactive oxygen and nitrogen species generated by atmospheric-pressure air plasma. PLoS ONE 2014, 9. [Google Scholar] [CrossRef]
- Gandhirajan, R.K.; Rödder, K.; Bodnar, Y.; Pasqual-Melo, G.; Emmert, S.; Griguer, C.E.; Weltmann, K.D.; Bekeschus, S. Cytochrome C oxidase Inhibition and Cold Plasma-derived Oxidants Synergize in Melanoma Cell Death Induction. Sci. Rep. 2018, 8, 12734. [Google Scholar] [CrossRef]
- Adachi, T.; Tanaka, H.; Nonomura, S.; Hara, H.; Kondo, S.I.; Hori, M. Plasma-activated medium induces A549 cell injury via a spiral apoptotic cascade involving the mitochondrial-nuclear network. Free Radic. Biol. Med. 2015, 79, 28–44. [Google Scholar] [CrossRef]
- Zhunussova, A.; Vitol, E.A.; Polyak, B.; Tuleukhanov, S.; Brooks, A.D.; Sensenig, R.; Friedman, G.; Orynbayeva, Z. Mitochondria-Mediated Anticancer Effects of Non-Thermal Atmospheric Plasma. PLoS ONE 2016, 11, e0156818. [Google Scholar] [CrossRef] [Green Version]
- Gjika, E.; Pal-Ghosh, S.; Tang, A.; Kirschner, M.; Tadvalkar, G.; Canady, J.; Stepp, M.A.; Keidar, M. Adaptation of Operational Parameters of Cold Atmospheric Plasma for in Vitro Treatment of Cancer Cells. ACS Appl. Mater. Interfaces 2018, 10, 9269–9279. [Google Scholar] [CrossRef]
- Tuhvatulin, A.I.; Sysolyatina, E.V.; Scheblyakov, D.V.; Logunov, D.Y.; Vasiliev, M.M.; Yurova, M.A.; Danilova, M.A.; Petrov, O.F.; Naroditsky, B.S.; Morfill, G.E.; et al. Non-thermal Plasma Causes p53-Dependent Apoptosis in Human Colon Carcinoma Cells. Acta Naturae 2012, 4, 82–87. [Google Scholar]
- Mariño, G.; Niso-Santano, M.; Baehrecke, E.H.; Kroemer, G. Self-consumption: The interplay of autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 2014, 15, 81–94. [Google Scholar] [CrossRef] [Green Version]
- Kumari, S.; Badana, A.K.; Mohan, M.; Shailender, G.; Malla, R. Reactive Oxygen Species: A Key Constituent in Cancer Survival. Biomark. Insights 2018, 13. [Google Scholar] [CrossRef] [Green Version]
- Girard, P.-M.; Arbabian, A.; Fleury, M.; Bauville, G.; Puech, V.; Dutreix, M.; Sousa, J.S. Synergistic Effect of H2O2 and NO2 in Cell Death Induced by Cold Atmospheric He Plasma. Sci. Rep. 2016, 6, 29098. [Google Scholar] [CrossRef] [Green Version]
- 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 (Review). Biointerphases Appl. Phys. Lett. 2015, 10, 40801–224101. [Google Scholar] [CrossRef] [Green Version]
- Bauer, G. The synergistic effect between hydrogen peroxide and nitrite, two long-lived molecular species from cold atmospheric plasma, triggers tumor cells to induce their own cell death. Redox Biol. 2019, 26. [Google Scholar] [CrossRef]
- Hosokawa, Y.; Saga, R.; Monzen, S.; Terashima, S.; Tsuruga, E. Ascorbic acid does not reduce the anticancer effect of radiotherapy. Biomed. Rep. 2017, 6, 103–107. [Google Scholar] [CrossRef] [Green Version]
- Ibañez, I.L.; Notcovich, C.; Catalano, P.N.; Bellino, M.G.; Durán, H. The redox-active nanomaterial toolbox for cancer therapy. Cancer Lett. 2015, 359, 9–19. [Google Scholar] [CrossRef]
- Semmler, M.L.; Bekeschus, S.; Schäfer, M.; Bernhardt, T.; Fischer, T.; Witzke, K.; Seebauer, C.; Rebl, H.; Grambow, E.; Vollmar, B.; et al. Molecular mechanisms of the efficacy of cold atmospheric pressure plasma (CAP) in cancer treatment. Cancers 2020, 12, 269. [Google Scholar]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Fan, T.M.; Roberts, R.D.; Lizardo, M.M. Understanding and Modeling Metastasis Biology to Improve Therapeutic Strategies for Combating Osteosarcoma Progression. Front. Oncol. 2020, 10, 13. [Google Scholar]
- Hirst, A.M.; Frame, F.M.; Maitland, N.J.; O’Connell, D. Low Temperature Plasma: A Novel Focal Therapy for Localized Prostate Cancer? Biomed. Res. Int. 2014, 2014. [Google Scholar] [CrossRef] [Green Version]
- Bogaerts, A.; Yusupov, M.; Razzokov, J.; Van der Paal, J. Plasma for cancer treatment: How can RONS penetrate through the cell membrane? Answers from computer modeling. Front. Chem. Sci. Eng. 2019, 13, 253–263. [Google Scholar]
- Griseti, E.; Kolosnjaj-Tabi, J.; Gibot, L.; Fourquaux, I.; Rols, M.-P.; Yousfi, M.; Merbahi, N.; Golzio, M. Pulsed Electric Field Treatment Enhances the Cytotoxicity of Plasma-Activated Liquids in a Three-Dimensional Human Colorectal Cancer Cell Model. Sci. Rep. 2019, 9, 7583. [Google Scholar] [CrossRef]
- Yu, H.; Wang, Y.; Wang, S.; Li, X.; Li, W.; Ding, D.; Gong, X.; Keidar, M.; Zhang, W. Paclitaxel-Loaded Core-Shell Magnetic Nanoparticles and Cold Atmospheric Plasma Inhibit Non-Small Cell Lung Cancer Growth. ACS Appl. Mater. Interfaces 2018, 10, 43462–43471. [Google Scholar] [CrossRef]
- Li, W.; Yu, H.; Ding, D.; Chen, Z.; Wang, Y.; Wang, S.; Li, X.; Keidar, M.; Zhang, W. Cold atmospheric plasma and iron oxide-based magnetic nanoparticles for synergetic lung cancer therapy. Free Radic. Biol. Med. 2019, 130, 71–81. [Google Scholar] [CrossRef]
- Privat-Maldonado, A.; Bengtson, C.; Razzokov, J.; Smits, E.; Bogaerts, A. Modifying the tumour microenvironment: Challenges and future perspectives for anticancer plasma treatments. Cancers 2019, 11, 1920. [Google Scholar]
- Freund, E.; Liedtke, K.R.; van der Linde, J.; Metelmann, H.R.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. Physical plasma-treated saline promotes an immunogenic phenotype in CT26 colon cancer cells in vitro and in vivo. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef]
- Tanaka, H.; Nakamura, K.; Mizuno, M.; Ishikawa, K.; Takeda, K.; Kajiyama, H.; Utsumi, F.; Kikkawa, F.; Hori, M. Non-thermal atmospheric pressure plasma activates lactate in Ringer’s solution for anti-tumor effects. Sci. Rep. 2016, 6, 36282. [Google Scholar] [CrossRef] [Green Version]
- Utsumi, F.; Kajiyama, H.; Nakamura, K.; Tanaka, H.; Mizuno, M.; Ishikawa, K.; Kondo, H.; Kano, H.; Hori, M.; Kikkawa, F. 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. [Google Scholar] [CrossRef] [Green Version]
- Kaushik, N.N.K.; Ghimire, B.; Li, Y.; Adhikari, M.; Veerana, M.; Kaushik, N.N.K.; Jha, N.; Adhikari, B.; Lee, S.-J.; Masur, K.; et al. Biological and medical applications of plasma-activated media, water and solutions. Biol. Chem. 2018, 400, 39–62. [Google Scholar] [CrossRef]
- Tornin, J.; Mateu-Sanz, M.; Rodríguez, A.; Labay, C.; Rodríguez, R.; Canal, C. Pyruvate Plays a Main Role in the Antitumoral Selectivity of Cold Atmospheric Plasma in Osteosarcoma. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [Green Version]
- Liedtke, K.R.; Freund, E.; Hermes, M.; Oswald, S.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. Gas plasma-conditioned ringer’s lactate enhances the cytotoxic activity of cisplatin and gemcitabine in pancreatic cancer in vitro and in ovo. Cancers 2020, 12, 123. [Google Scholar] [CrossRef] [Green Version]
- He, Z.; Liu, K.; Manaloto, E.; Casey, A.; Cribaro, G.P.; Byrne, H.J.; Tian, F.; Barcia, C.; Conway, G.E.; Cullen, P.J.; et al. Cold Atmospheric Plasma Induces ATP-Dependent Endocytosis of Nanoparticles and Synergistic U373MG Cancer Cell Death. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
- Shaw, P.; Kumar, N.; Hammerschmid, D.; Privat-Maldonado, A.; Dewilde, S.; Bogaerts, A. Synergistic Effects of Melittin and Plasma Treatment: A Promising Approach for Cancer Therapy. Cancers 2019, 11, 1109. [Google Scholar] [CrossRef] [Green Version]
- Narayanaswamy, R.; Torchilin, V.P. Hydrogels and their applications in targeted drug delivery. Molecules 2019, 24, 603. [Google Scholar]
- Liu, M.; Zeng, X.; Ma, C.; Yi, H.; Ali, Z.; Mou, X.; Li, S.; Deng, Y.; He, N. Injectable hydrogels for cartilage and bone tissue engineering. Bone Res. 2017, 5, 17014. [Google Scholar]
- Bai, X.; Gao, M.; Syed, S.; Zhuang, J.; Xu, X.; Zhang, X.Q. Bioactive hydrogels for bone regeneration. Bioact. Mater. 2018, 3, 401–417. [Google Scholar]
- Labay, C.; Hamouda, I.; Tampieri, F.; Ginebra, M.-P.; Canal, C. Production of reactive species in alginate hydrogels for cold atmospheric plasma-based therapies. Sci. Rep. 2019, 9, 16160. [Google Scholar] [CrossRef]
- Canal, C.; Fontelo, R.; Hamouda, I.; Guillem-Marti, J.; Cvelbar, U.; Ginebra, M.P. Plasma-induced selectivity in bone cancer cells death. Free Radic. Biol. Med. 2017, 110, 72–80. [Google Scholar] [CrossRef] [Green Version]
- Gümbel, D.; Suchy, B.; Wien, L.; Gelbrich, N.; Napp, M.; Kramer, A.; Ekkernkamp, A.; Daeschlein, G.; Stope, M.B. Comparison of Cold Atmospheric Plasma Devices’ Efficacy on Osteosarcoma and Fibroblastic In Vitro Cell Models. Anticancer Res. 2017, 37, 5407–5414. [Google Scholar] [CrossRef]
- Haralambiev, L.; Wien, L.; Gelbrich, N.; Lange, J.; Bakir, S.; Kramer, A.; Burchardt, M.; Ekkernkamp, A.; Gümbel, D.; Stope, M.B. Cold atmospheric plasma inhibits the growth of osteosarcoma cells by inducing apoptosis, independent of the device used. Oncol. Lett. 2020, 19, 283–290. [Google Scholar] [CrossRef] [Green Version]
- Gümbel, D.; Gelbrich, N.; Weiss, M.; Napp, M.; Daeschlein, G.; Sckell, A.; Ender, S.A.; Kramer, A.; Burchardt, M.; Ekkernkamp, A.; et al. New Treatment Options for Osteosarcoma—Inactivation of Osteosarcoma Cells by Cold Atmospheric Plasma. Anticancer Res. 2016, 36, 5915–5922. [Google Scholar] [CrossRef] [Green Version]
- Gümbel, D.; Gelbrich, N.; Napp, M.; Daeschlein, G.; Kramer, A.; Sckell, A.; Burchardt, M.; Ekkernkamp, A.; Stope, M.B. Peroxiredoxin Expression of Human Osteosarcoma Cells Is Influenced by Cold Atmospheric Plasma Treatment. Anticancer Res. 2017, 37, 1031–1038. [Google Scholar] [CrossRef] [Green Version]
- Haralambiev, L.; Wien, L.; Gelbrich, N.; Kramer, A.; Mustea, A.; Burchardt, M.; Ekkernkamp, A.; Stope, M.B.; Gümbel, D. Effects of Cold Atmospheric Plasma on the Expression of Chemokines, Growth Factors, TNF Superfamily Members, Interleukins, and Cytokines in Human Osteosarcoma Cells. Anticancer Res. 2019, 39, 151–157. [Google Scholar] [CrossRef]
- Haralambiev, L.; Nitsch, A.; Einenkel, R.; Muzzio, D.O.; Gelbrich, N.; Burchardt, M.; Zygmunt, M.; Ekkernkamp, A.; Stope, M.B.; Gümbel, D. The effect of cold atmospheric plasma on the membrane permeability of human osteosarcoma cells. Anticancer Res. 2020, 40, 841–846. [Google Scholar] [CrossRef]
- Jacoby, J.M.; Strakeljahn, S.; Nitsch, A.; Bekeschus, S.; Hinz, P.; Mustea, A.; Ekkernkamp, A.; Tzvetkov, M.V.; Haralambiev, L.; Stope, M.B. An Innovative Therapeutic Option for the Treatment of Skeletal Sarcomas: Elimination of Osteo- and Ewing’s Sarcoma Cells Using Physical Gas Plasma. Int. J. Mol. Sci. 2020, 12, 4460. [Google Scholar]
- Ito, T.; Ando, T.; Suzuki-Karasaki, M.; Tokunaga, T.; Yoshida, Y.; Ochiai, T.; Tokuhashi, Y.; Suzuki-Karasaki, Y. Cold PSM, but not TRAIL, triggers autophagic cell death: A therapeutic advantage of PSM over TRAIL. Int. J. Oncol. 2018, 53, 503–514. [Google Scholar] [CrossRef]
- Tokunaga, T.; Ando, T.; Suzuki-Karasaki, M.; Ito, T.; Onoe-Takahashi, A.; Ochiai, T.; Soma, M.; Suzuki-Karasaki, Y. Plasma-stimulated medium kills TRAIL-resistant human malignant cells by promoting caspase-independent cell death via membrane potential and calcium dynamics modulation. Int. J. Oncol. 2018, 52, 697–708. [Google Scholar] [CrossRef] [Green Version]
- Mateu-Sanz, M.; Tornin, J.; Brulin, B.; Khlyustova, A.; Ginebra, M.-P.; Layrolle, P.; Canal, C. Cold Plasma-Treated Ringer’s Saline: A Weapon to Target Osteosarcoma. Cancers 2020, 12, 227. [Google Scholar] [CrossRef] [Green Version]
- Chung, T.-H.; Stancampiano, A.; Sklias, K.; Gazeli, K.; André, F.; Dozias, S.; Douat, C.; Pouvesle, J.-M.; Santos Sousa, J.; Robert, É.; et al. Cell Electropermeabilisation Enhancement by Non-Thermal-Plasma-Treated PBS. Cancers 2020, 12, 219. [Google Scholar] [CrossRef] [Green Version]
- Li, P.; Shi, X.; Xu, Y.; Zhong, B.; Lu, Y.; Sun, Y. Interleukin-22 promotes osteosarcoma cell proliferation and invasion via STAT3 activation. Med. Sci. Monit. 2018, 24, 7802–7808. [Google Scholar] [CrossRef]
- Yang, J.; Yang, D.; Sun, Y.; Sun, B.; Wang, G.; Trent, J.C.; Araujo, D.M.; Chen, K.; Zhang, W. Genetic amplification of the vascular endothelial growth factor (VEGF) pathway genes, including VEGFA, in human osteosarcoma. Cancer 2011, 117, 4925–4938. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.J.; Chung, T.H. Cold atmospheric plasma jet-generated RONS and their selective effects on normal and carcinoma cells. Sci. Rep. 2016, 6, 20332. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.-Y.; Wu, P.-K.; Chen, P.C.-H.; Lee, C.-W.; Chen, W.-M.; Hung, S.-C. Generation of Osteosarcomas from a Combination of Rb Silencing and c-Myc Overexpression in Human Mesenchymal Stem Cells. Stem Cells Transl. Med. 2017, 6, 512–526. [Google Scholar] [CrossRef] [Green Version]
- Peng, C.; Yang, Q.; Wei, B.; Yuan, B.; Liu, Y.; Li, Y.; Gu, D.; Yin, G.; Wang, B.; Xu, D.; et al. Investigation of crucial genes and microRNAs in conventional osteosarcoma using gene expression profling analysis. Mol. Med. Rep. 2017, 16, 7617–7624. [Google Scholar] [CrossRef]
- Xiao, W.; Mohseny, A.B.; Hogendoorn, P.C.W.; Cleton-Jansen, A.-M. Mesenchymal stem cell transformation and sarcoma genesis. Clin. Sarcoma Res. 2013, 3, 10. [Google Scholar] [CrossRef] [Green Version]
- Shen, H.; Wang, W.; Ni, B.; Zou, Q.; Lu, H.; Wang, Z. Exploring the molecular mechanisms of osteosarcoma by the integrated analysis of mRNAs and miRNA microarrays. Int. J. Mol. Med. 2018, 42, 21–30. [Google Scholar] [CrossRef]
- Zhao, C.; Zhang, Q.; Yu, T.; Sun, S.; Wang, W.; Liu, G. Hypoxia promotes drug resistance in osteosarcoma cells via activating AMP-activated protein kinase (AMPK) signaling. J. Bone Oncol. 2016, 5, 22–29. [Google Scholar] [CrossRef] [Green Version]
- Ren, K.; Lu, X.; Yao, N.; Chen, Y.; Yang, A.; Chen, H.; Zhang, J.; Wu, S.; Shi, X.; Wang, C.; et al. Focal adhesion kinase overexpression and its impact on human osteosarcoma. Oncotarget 2015, 6, 31085–31103. [Google Scholar] [CrossRef]
- Van der Worp, H.B.; Howells, D.W.; Sena, E.S.; Porritt, M.J.; Rewell, S.; O’Collins, V.; Macleod, M.R. Can Animal Models of Disease Reliably Inform Human Studies? PLoS Med. 2010, 7, e1000245. [Google Scholar] [CrossRef] [Green Version]
- De Luca, A.; Raimondi, L.; Salamanna, F.; Carina, V.; Costa, V.; Bellavia, D.; Alessandro, R.; Fini, M.; Giavaresi, G. Relevance of 3d culture systems to study osteosarcoma environment. J. Exp. Clin. Cancer Res. 2018, 37, 1–15. [Google Scholar]
- Monteiro, C.F.; Custódio, C.A.; Mano, J.F. Three-Dimensional Osteosarcoma Models for Advancing Drug Discovery and Development. Adv. Ther. 2019, 2, 1800108. [Google Scholar] [CrossRef]
- Alfranca, A.; Martinez-Cruzado, L.; Tornin, J.; Abarrategi, A.; Amaral, T.; De Alava, E.; Menendez, P.; Garcia-Castro, J.; Rodriguez, R. Bone microenvironment signals in osteosarcoma development. Cell. Mol. Life Sci. 2015, 72, 3097–3113. [Google Scholar]
- Brown, H.K.; Tellez-Gabriel, M.; Heymann, D. Cancer stem cells in osteosarcoma. Cancer Lett. 2017, 386, 189–195. [Google Scholar] [CrossRef]
- Rubio, R.; Abarrategi, A.; Garcia-Castro, J.; Martinez-Cruzado, L.; Suarez, C.; Tornin, J.; Santos, L.; Astudillo, A.; Colmenero, I.; Mulero, F.; et al. Bone Environment is Essential for Osteosarcoma Development from Transformed Mesenchymal Stem Cells. Stem Cells 2014, 32, 1136–1148. [Google Scholar] [CrossRef]
- Navet, B.; Ando, K.; Vargas-Franco, J.W.; Brion, R.; Amiaud, J.; Mori, K.; Yagita, H.; Mueller, C.G.; Verrecchia, F.; Dumars, C.; et al. The intrinsic and extrinsic implications of RANKL/RANK signaling in osteosarcoma: From tumor initiation to lung metastases. Cancers 2018, 10, 398. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.S.; Liu, Q.; Tian, J.; He, H.B.; Luo, W. Angiogenesis Process in Osteosarcoma: An Updated Perspective of Pathophysiology and Therapeutics. Am. J. Med. Sci. 2019, 357, 280–288. [Google Scholar]
- Tu, B.; Du, L.; Fan, Q.M.; Tang, Z.; Tang, T.T. STAT3 activation by IL-6 from mesenchymal stem cells promotes the proliferation and metastasis of osteosarcoma. Cancer Lett. 2012, 325, 80–88. [Google Scholar] [CrossRef]
- Bian, Z.Y.; Fan, Q.M.; Li, G.; Xu, W.T.; Tang, T.T. Human mesenchymal stem cells promote growth of osteosarcoma: Involvement of interleukin-6 in the interaction between human mesenchymal stem cells and Saos-2. Cancer Sci. 2010, 101, 2554–2560. [Google Scholar] [CrossRef]
- Tsukamoto, S.; Honoki, K.; Fuji, H.; Tohma, Y.; Kido, A.; Mori, T.; Tsujiuchi, T.; Tanaka, Y. Mesenchymal stem cells promote tumor engraftment and metastatic colonization in rat osteosarcoma model. Int. J. Oncol. 2012, 40, 163–169. [Google Scholar] [CrossRef]
- Zhang, P.; Dong, L.; Long, H.; Yang, T.T.; Zhou, Y.; Fan, Q.Y.; Ma, B.A. Homologous mesenchymal stem cells promote the emergence and growth of pulmonary metastases of the rat osteosarcoma cell line UMR-106. Oncol. Lett. 2014, 8, 127–132. [Google Scholar] [CrossRef]
- Zheng, Y.; Wang, G.; Chen, R.; Hua, Y.; Cai, Z. Mesenchymal stem cells in the osteosarcoma microenvironment: Their biological properties, influence on tumor growth, and therapeutic implications. Stem Cell Res. Ther. 2018, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Cortini, M.; Avnet, S.; Baldini, N. Mesenchymal stroma: Role in osteosarcoma progression. Cancer Lett. 2017, 405, 90–99. [Google Scholar] [CrossRef]
- Birru, B.; Durthi, C.P.; Kacham, S.; Pola, M.; Rajulapati, S.B.; Parcha, S.R.; Kamal, M.A. Stem Cells in Tumour Microenvironment Aids for Prolonged Survival Rate of Cancer Cells and Developed Drug Resistance: Major Challenge in Osteosarcoma Treatment. Curr. Drug Metab. 2020, 21. [Google Scholar] [CrossRef]
- Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar]
- Koirala, P.; Roth, M.E.; Gill, J.; Piperdi, S.; Chinai, J.M.; Geller, D.S.; Hoang, B.H.; Park, A.; Fremed, M.A.; Zang, X.; et al. Immune infiltration and PD-L1 expression in the tumor microenvironment are prognostic in osteosarcoma. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef]
- Park, J.; Lee, H.; Lee, H.J.; Kim, G.C.; Kim, S.S.; Han, S.; Song, K. Non-thermal atmospheric pressure plasma is an excellent tool to activate proliferation in various mesoderm-derived human adult stem cells. Free Radic. Biol. Med. 2019, 134, 374–384. [Google Scholar] [CrossRef]
- Chernets, N.; Zhang, J.; Steinbeck, M.J.; Kurpad, D.S.; Koyama, E.; Friedman, G.; Freeman, T.A. Nonthermal atmospheric pressure plasma enhances mouse limb bud survival, growth, and elongation. Tissue Eng. Part A 2015, 21, 300–309. [Google Scholar] [CrossRef] [Green Version]
- Steinbeck, M.J.; Chernets, N.; Zhang, J.; Kurpad, D.S.; Fridman, G.; Fridman, A.; Freeman, T.A. Skeletal cell differentiation is enhanced by atmospheric dielectric barrier discharge plasma treatment. PLoS ONE 2013, 8, e82143. [Google Scholar] [CrossRef]
- Tominami, K.; Kanetaka, H.; Sasaki, S.; Mokudai, T.; Kaneko, T.; Niwano, Y. Cold atmospheric plasma enhances osteoblast differentiation. PLoS ONE 2017, 12, e0180507. [Google Scholar] [CrossRef]
- Lin, A.; Truong, B.; Patel, S.; Kaushik, N.; Choi, E.H.; Fridman, G.; Fridman, A.; Miller, V. Nanosecond-Pulsed DBD Plasma-Generated Reactive Oxygen Species Trigger Immunogenic Cell Death in A549 Lung Carcinoma Cells through Intracellular Oxidative Stress. Int. J. Mol. Sci. 2017, 18, 966. [Google Scholar] [CrossRef] [Green Version]
- Miao, X.; Yin, S.; Shao, Z.; Zhang, Y.; Chen, X. Nanosecond pulsed electric field inhibits proliferation and induces apoptosis in human osteosarcoma. J. Orthop. Surg. Res. 2015, 10. [Google Scholar] [CrossRef] [Green Version]
- Gweon, B.; Kim, H.; Kim, K.; Kim, M.; Shim, E.; Kim, S.; Choe, W.; Shin, J.H. Suppression of angiogenesis by atmospheric pressure plasma in human aortic endothelial cells. Appl. Phys. Lett. 2014, 104, 133701. [Google Scholar] [CrossRef] [Green Version]
- Virard, F.; Cousty, S.; Cambus, J.-P.; Valentin, A.; Kémoun, P.; Clément, F. Cold Atmospheric Plasma Induces a Predominantly Necrotic Cell Death via the Microenvironment. PLoS ONE 2015, 10, e0133120. [Google Scholar] [CrossRef]
- Bonuccelli, G.; Avnet, S.; Grisendi, G.; Salerno, M.; Granchi, D.; Dominici, M.; Kusuzaki, K.; Baldini, N. Role of mesenchymal stem cells in osteosarcoma and metabolic reprogramming of tumor cells. Oncotarget 2014, 5, 7575–7588. [Google Scholar] [CrossRef]
- Liedtke, K.R.; Freund, E.; Hackbarth, C.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. A myeloid and lymphoid infiltrate in murine pancreatic tumors exposed to plasma-treated medium. Clin. Plasma Med. 2018, 11, 10–17. [Google Scholar] [CrossRef]
- Van Loenhout, J.; Flieswasser, T.; Boullosa, L.F.; De Waele, J.; Van Audenaerde, J.; Marcq, E.; Jacobs, J.; Lin, A.; Lion, E.; Dewitte, H.; et al. Cold atmospheric plasma-treated PBS eliminates immunosuppressive pancreatic stellate cells and induces immunogenic cell death of pancreatic cancer cells. Cancers 2019, 11, 597. [Google Scholar] [CrossRef] [Green Version]
- Yao, D.; Cai, G.-H.; Chen, J.; Ling, R.; Wu, S.-X.; Li, Y.-P. Prognostic value of p53 alterations in human osteosarcoma: A meta analysis. Int. J. Clin. Exp. Pathol. 2014, 7, 6725–6733. [Google Scholar]
- Velletri, T.; Xie, N.; Wang, Y.; Huang, Y.; Yang, Q.; Chen, X.; Chen, Q.; Shou, P.; Gan, Y.; Cao, G.; et al. P53 functional abnormality in mesenchymal stem cells promotes osteosarcoma development. Cell Death Dis. 2016, 7, e2015. [Google Scholar]
- Lu, Y.; Gitelis, S.; Lei, G.; Ding, M.; Maki, C.; Mira, R.R.; Zheng, Q. Research findings working with the p53 and Rb1 targeted osteosarcoma mouse model. Am. J. Cancer Res. 2014, 4, 234–244. [Google Scholar]
- Li, J.; Yang, Z.; Li, Y.; Xia, J.; Li, D.; Li, H.; Ren, M.; Liao, Y.; Yu, S.; Chen, Y.; et al. Cell apoptosis, autophagy and necroptosis in osteosarcoma treatment. Oncotarget 2016, 7, 44763–44778. [Google Scholar]
- Porta, C.; Paglino, C.; Mosca, A. Targeting PI3K/Akt/mTOR signaling in cancer. Front. Oncol. 2014, 4, 64. [Google Scholar]
- Chen, B.J.; Wu, Y.L.; Tanaka, Y.; Zhang, W. Small molecules targeting c-Myc oncogene: Promising anti-cancer therapeutics. Int. J. Biol. Sci. 2014, 10, 1084–1096. [Google Scholar] [CrossRef]
- Wang, Q.; Liu, H.; Wang, Q.; Zhou, F.; Liu, Y.; Zhang, Y.; Ding, H.; Yuan, M.; Li, F.; Chen, Y. Involvement of c-Fos in cell proliferation, migration, and invasion in osteosarcoma cells accompanied by altered expression of Wnt2 and Fzd9. PLoS ONE 2017, 12. [Google Scholar] [CrossRef] [Green Version]
- Poos, K.; Smida, J.; Maugg, D.; Eckstein, G.; Baumhoer, D.; Nathrath, M.; Korsching, E. Genomic heterogeneity of osteosarcoma—Shift from single candidates to functional modules. PLoS ONE 2015, 10. [Google Scholar] [CrossRef] [Green Version]
- Xu, W.; Xu, J.; Wang, T.; Liu, W.; Wei, H.; Yang, X.; Yan, W.; Zhou, W.; Xiao, J. Ellagic acid and sennoside B inhibit osteosarcoma cell migration, invasion and growth by repressing the expression of c-jun. Oncol. Lett. 2018, 16, 898–904. [Google Scholar] [CrossRef]
- Schmidt, A.; Bekeschus, S.; Jarick, K.; Hasse, S.; von Woedtke, T.; Wende, K. Cold Physical Plasma Modulates p53 and Mitogen-Activated Protein Kinase Signaling in Keratinocytes. Oxid. Med. Cell. Longev. 2019, 2019, 7017363. [Google Scholar] [CrossRef] [Green Version]
- Shi, L.; Yu, L.; Zou, F.; Hu, H.; Liu, K.; Lin, Z. Gene expression profiling and functional analysis reveals that p53 pathway-related gene expression is highly activated in cancer cells treated by cold atmospheric plasma-activated medium. PeerJ 2017, 5, e3751. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Bahrami, A.; Pappo, A.; Easton, J.; Dalton, J.; Hedlund, E.; Ellison, D.; Shurtleff, S.; Wu, G.; Wei, L.; et al. Recurrent Somatic Structural Variations Contribute to Tumorigenesis in Pediatric Osteosarcoma. Cell Rep. 2014, 7, 104. [Google Scholar] [CrossRef] [Green Version]
- Leroy, B.; Girard, L.; Hollestelle, A.; Minna, J.D.; Gazdar, A.F.; Soussi, T. Analysis of TP53 mutation status in human cancer cell lines: A reassessment. Hum. Mutat. 2014, 35, 756–765. [Google Scholar] [CrossRef]
- Bundscherer, L.; Wende, K.; Ottmüller, K.; Barton, A.; Schmidt, A.; Bekeschus, S.; Hasse, S.; Weltmann, K.D.; Masur, K.; Lindequist, U. Impact of non-thermal plasma treatment on MAPK signaling pathways of human immune cell lines. Immunobiology 2013, 218, 1248–1255. [Google Scholar] [CrossRef]
- Kumar, N.; Attri, P.; Yadav, D.K.; Choi, J.; Choi, E.H.; Uhm, H.S. Induced apoptosis in melanocytes cancer cell and oxidation in biomolecules through deuterium oxide generated from atmospheric pressure non-thermal plasma jet. Sci. Rep. 2014, 4. [Google Scholar] [CrossRef] [Green Version]
- Xia, J.; Zeng, W.; Xia, Y.; Wang, B.; Xu, D.; Liu, D.; Kong, M.G.; Dong, Y. Cold atmospheric plasma induces apoptosis of melanoma cells via Sestrin2-mediated nitric oxide synthase signaling. J. Biophotonics 2019, 12. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, K.; Peng, Y.; Utsumi, F.; Tanaka, H.; Mizuno, M.; Toyokuni, S.; Hori, M.; Kikkawa, F.; Kajiyama, H. Novel Intraperitoneal Treatment With Non-Thermal Plasma-Activated Medium Inhibits Metastatic Potential of Ovarian Cancer Cells. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef]
- Tang, Z.; Zhao, L.; Yang, Z.; Liu, Z.; Gu, J.; Bai, B.; Liu, J.; Xu, J.; Yang, H. Mechanisms of oxidative stress, apoptosis, and autophagy involved in graphene oxide nanomaterial anti-osteosarcoma effect. Int. J. Nanomedicine 2018, 13, 2907–2919. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Wang, W.; Qiu, E. Protection of oxidative stress induced apoptosis in osteosarcoma cells by dihydromyricetin through down-regulation of caspase activation and up-regulation of BcL-2. Saudi J. Biol. Sci. 2017, 24, 837–842. [Google Scholar] [CrossRef]
- Mutsaers, A.J.; Walkley, C.R. Cells of origin in osteosarcoma: Mesenchymal stem cells or osteoblast committed cells? Bone 2014, 62, 56–63. [Google Scholar] [CrossRef]
- Abarrategi, A.; Tornin, J.; Lucia, M.C.; Hamilton, A.; Enrique, M.C.; Rodrigo, J.P.; González, M.V.; Baldini, N.; Javier, G.C.; Rodriguez, R. Osteosarcoma: Cells-of-Origin, Cancer stem cells, and targeted therapies. Stem Cells Int. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
- Shi, X.; Zhang, Y.; Zheng, J.; Pan, J. Reactive Oxygen Species in Cancer Stem Cells. Antioxid. Redox Signal. 2012, 16, 1215–1228. [Google Scholar] [CrossRef] [Green Version]
- Abdal Dayem, A.; Choi, H.Y.; Kim, J.H.; Cho, S.G. Role of oxidative stress in stem, cancer, and cancer stem cells. Cancers 2010, 2, 859–884. [Google Scholar]
- Ikeda, J.; Tanaka, H.; Ishikawa, K.; Sakakita, H.; Ikehara, Y.; Hori, M. Plasma-activated medium (PAM) kills human cancer-initiating cells. Pathol. Int. 2018, 68, 23–30. [Google Scholar] [CrossRef]
- Ikeda, J.-I. Effect of Nonequilibrium Atmospheric Pressure Plasma on Cancer-Initiating Cells. Plasma Med. 2014, 4, 49–56. [Google Scholar]
- Kurake, N.; Ishikawa, K.; Tanaka, H.; Hashizume, H.; Nakamura, K.; Kajiyama, H.; Toyokuni, S.; Kikkawa, F.; Mizuno, M.; Hori, M. Non-thermal plasma-activated medium modified metabolomic profiles in the glycolysis of U251SP glioblastoma. Arch. Biochem. Biophys. 2019, 662, 83–92. [Google Scholar] [CrossRef]
- Schmidt, A.; Wende, K.; Bekeschus, S.; Bundscherer, L.; Barton, A.; Ottmüller, K.; Weltmann, K.D.; Masur, K. Non-thermal plasma treatment is associated with changes in transcriptome of human epithelial skin cells. Free Radic. Res. 2013, 47, 577–592. [Google Scholar] [CrossRef]
- Yoshida, G.J.; Saya, H. Therapeutic strategies targeting cancer stem cells. Cancer Sci. 2016, 107, 5–11. [Google Scholar]
- Funes, J.M.; Quintero, M.; Henderson, S.; Martinez, D.; Qureshi, U.; Westwood, C.; Clements, M.O.; Bourboulia, D.; Pedley, R.B.; Moncada, S.; et al. Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proc. Natl. Acad. Sci. USA 2007, 104, 6223–6228. [Google Scholar] [CrossRef] [Green Version]
- Depeng, S.; Wu, J.; Guo, L.; Xu, Y.; Liu, L.; Lu, J. Metformin increases sensitivity of osteosarcoma stem cells to cisplatin by inhibiting expression of PKM2. Int. J. Oncol. 2017, 50, 1848–1856. [Google Scholar] [CrossRef]
- Chandhanayingyong, C.; Kim, Y.; Staples, J.R.; Hahn, C.; Lee, F.Y. MAPK/ERK signaling in osteosarcomas, Ewing sarcomas and chondrosarcomas: Therapeutic implications and future directions. Sarcoma 2012, 2012. [Google Scholar] [CrossRef]
- Qi, X.T.; Li, Y.L.; Zhang, Y.Q.; Xu, T.; Lu, B.; Fang, L.; Gao, J.Q.; Yu, L.S.; Zhu, D.F.; Yang, B.; et al. KLF4 functions as an oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells. Acta Pharmacol. Sin. 2019, 40, 546–555. [Google Scholar] [CrossRef]
- Hu, X.; Wang, Z.; Chen, M.; Chen, X.; Liang, W. The anti-osteosarcoma cell activity by a mTORC1/2 dual inhibitor RES-529. Biochem. Biophys. Res. Commun. 2018, 497, 499–505. [Google Scholar] [CrossRef]
- Zeng, X.; Wang, S.; Gui, P.; Wu, H.; Li, Z. Expression and significance of Annexin A3 in the osteosarcoma cell lines HOS and U2OS. Mol. Med. Rep. 2019, 20, 2583–2590. [Google Scholar] [CrossRef]
- Li, P.C.; Tu, M.J.; Ho, P.Y.; Jilek, J.L.; Duan, Z.; Zhang, Q.Y.; Yu, A.X.; Yu, A.M. Bioengineered NRF2-siRNA is effective to interfere with NRF2 pathways and improve chemosensitivity of human cancer cells. Drug Metab. Dispos. 2018, 46, 2–10. [Google Scholar] [CrossRef] [Green Version]
- Zuo, D.; Shogren, K.L.; Zang, J.; Jewison, D.E.; Waletzki, B.E.; Miller, A.L.; Okuno, S.H.; Cai, Z.; Yaszemski, M.J.; Maran, A. Inhibition of STAT3 blocks protein synthesis and tumor metastasis in osteosarcoma cells 06 Biological Sciences 0601 Biochemistry and Cell Biology. J. Exp. Clin. Cancer Res. 2018, 37, 244. [Google Scholar] [CrossRef] [Green Version]
- Zheng, B.; Ren, T.; Huang, Y.; Guo, W. Apatinib inhibits migration and invasion as well as PD-L1 expression in osteosarcoma by targeting STAT3. Biochem. Biophys. Res. Commun. 2017, 495, 1695–1701. [Google Scholar] [CrossRef]
- Pace, A.; Barone, G.; Lauria, A.; Martorana, A.; Palumbo Piccionello, A.; Pierro, P.; Terenzi, A.; Maria Almerico, A.; Buscemi, S.; Campanella, C.; et al. Hsp60, a Novel Target for Antitumor Therapy: Structure-Function Features and Prospective Drugs Design. Curr. Pharmaceut. Design 2013, 19, 2757–2764. [Google Scholar]
- Bekeschus, S.; Lippert, M.; Diepold, K.; Chiosis, G.; Seufferlein, T.; Azoitei, N. Physical plasma-triggered ROS induces tumor cell death upon cleavage of HSP90 chaperone. Sci. Rep. 2019, 9, 4112. [Google Scholar] [CrossRef] [Green Version]
- Marturano-Kruik, A.; Villasante, A.; Vunjak-Novakovic, G. Bioengineered models of solid human tumors for cancer research. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2016; Volume 1502, pp. 203–211. [Google Scholar]
- Takagi, S.; Takemoto, A.; Takami, M.; Oh-hara, T.; Fujita, N. Platelets promote osteosarcoma cell growth through activation of the platelet-derived growth factor receptor-Akt signaling axis. Cancer Sci. 2014, 105, 983–988. [Google Scholar] [CrossRef] [Green Version]
- Wang, F.; Schmidt, H.; Pavleska, D.; Wermann, T.; Seekamp, A.; Fuchs, S. Crude fucoidan extracts impair angiogenesis in models relevant for bone regeneration and osteosarcoma via reduction of VEGF and SDF-1. Mar. Drugs 2017, 15, 186. [Google Scholar] [CrossRef] [Green Version]
- Schiller, K.R.; Zillhardt, M.R.; Alley, J.; Borjesson, D.L.; Beitz, A.J.; Mauro, L.J. Secretion of MCP-1 and other paracrine factors in a novel tumor-bone coculture model. BMC Cancer 2009, 9, 45. [Google Scholar] [CrossRef] [Green Version]
BASIS | ADVANTAGES | DISADVANTAGES | |
---|---|---|---|
SURGERY | Limb amputation; Surgical resection of tumor tissue | ↑ Tumor remission and survival in non-metastatic OS patients. | ↑ Tumor residues, relapse and limb disfunction. ↓ Effectiveness in metastatic OS patients. |
CHEMO- THERAPY | Methotrexate, doxorubicin and cisplatin: inhibits DNA synthesis; Doxorubicin and cisplatin: Free radical production | ↓ Tumor growth; Tumor remission facilitates surgical resection; Eradicates tumor remnants and distal metastasis. | Drug resistance in many patients; Crystal nephropathy; Systemic oxidative stress: hepatotoxicity and cardiotoxicity; Hepatotoxicity, cardiotoxicity, altered bone remodeling function, side effects.any side effects. |
RADIOTHERAPY | Radiation-induced DNA damage; Production of hydroxyl radicals (·OH). | To control of resection margins; Local control of OS tumors that cannot be properly resected. | ↓ Response of OS tumors and a need for ↑ doses; Detrimental effect on normal tissue; Systemic oxidative stress and cytotoxicity; Risk of a radiation-induced secondary tumor. |
IMMUNOTHERAPY | Use of components of the immune system to increase the immune response against cancer cells. | ↓ Side effects than chemo- and radiotherapies and risk of tumor relapse. | ↑ Capabilities of OS to ignore immune system; Autoimmune responses. |
TARGETED THERAPIES | Use of different kinds of inhibitors of critical proto-oncogenes. | Targeted for OS cells; Free of systemic effects. | ↑ Difficulty to identify relevant proto-oncogenes in OS. |
Cell Lines | CAP Device | Cell Response | Refs |
---|---|---|---|
DIRECT CAP TREATMENT→ floating cultures except α | |||
SaOS-2, hMSCs hOBs α | He APPJ * | Cytotoxicity of cancer cells to CAP rather than healthy bone cells. | [112] |
U2-OS, 3T3 | Maxium® CAP Coagulator 1000 kINPen MED | Differential ↓ in proliferation depending on the plasma jet. | [113] |
U2-OS, MNNG/HOS | kINPen MED MiniJet-R | Plasma jet-dependent response; ↓ cell proliferation; activation of caspase-3/7. | [114] |
kINPen MED | ↓ Cell proliferation; p53 phosphorylation; DNA condensation and nuclear degradation. | [115] | |
↓ Cell proliferation and peroxiredoxin expression; NAC-mediated reduction of CAP cytotoxicity. | [116] | ||
Cell line-dependent chemokine and cytokine modulation. | [117] | ||
↑ Cell membrane permeability. | [118] | ||
↓ Cell proliferation and cell membrane permeability; apoptotic cell death. | [119] | ||
INDIRECT CAP TREATMENT (PLASMA-TREATED LIQUIDS) adherent cultures except β | |||
HOS, SaOS-2, 143B | DBD * | Mitochondrial network aberration, ↑ autophagy. | [120] |
HOS, SaOS-2, 143B, hFOB, LM8, K7M3, MC-3T3 | Cytotoxic effect in transformed cells; mitochondrial network aberration; caspase-independent cell death; cell membrane depolarization; Ca2+ homeostasis disruption. | [121] | |
SaOS-2, hMSCs, hOBs | He APPJ | ↑ Cytotoxicity of CAP to cancer cells than healthy bone cells and apoptosis; ↓ focal adhesions. | [112] |
SaOS-2, hBM-MSCs | Selective cytotoxic effects depending on H2O2 generated and the presence of pyruvate, ↑ DNA damage and apoptosis, phospho-kinase alterations. | [104] | |
SaOS-2, MG-63, U2-OS, hBM-MSCs | He APPJ kINPen IND | Selective cell death depending on plasma jet and RONS concentration, induction of intracellular ROS increase, DNA damage and apoptosis between healthy and cancer cells. | [122] |
Tumors produced from MOS-J β | He APPJ | ↓ Proliferating cells and viability. | [122] |
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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mateu-Sanz, M.; Tornín, J.; Ginebra, M.-P.; Canal, C. Cold Atmospheric Plasma: A New Strategy Based Primarily on Oxidative Stress for Osteosarcoma Therapy. J. Clin. Med. 2021, 10, 893. https://doi.org/10.3390/jcm10040893
Mateu-Sanz M, Tornín J, Ginebra M-P, Canal C. Cold Atmospheric Plasma: A New Strategy Based Primarily on Oxidative Stress for Osteosarcoma Therapy. Journal of Clinical Medicine. 2021; 10(4):893. https://doi.org/10.3390/jcm10040893
Chicago/Turabian StyleMateu-Sanz, Miguel, Juan Tornín, Maria-Pau Ginebra, and Cristina Canal. 2021. "Cold Atmospheric Plasma: A New Strategy Based Primarily on Oxidative Stress for Osteosarcoma Therapy" Journal of Clinical Medicine 10, no. 4: 893. https://doi.org/10.3390/jcm10040893
APA StyleMateu-Sanz, M., Tornín, J., Ginebra, M. -P., & Canal, C. (2021). Cold Atmospheric Plasma: A New Strategy Based Primarily on Oxidative Stress for Osteosarcoma Therapy. Journal of Clinical Medicine, 10(4), 893. https://doi.org/10.3390/jcm10040893