262
Views
1
CrossRef citations to date
0
Altmetric
Review Article

Current understanding of plasma-activated solutions for potential cancer therapy

ORCID Icon, & ORCID Icon
Pages 69-80 | Received 28 Dec 2022, Accepted 14 Mar 2023, Published online: 28 Mar 2023

References

  • Tendero C, Tixier C, Tristant P, et al. Atmospheric pressure plasmas: a review. Spectrochim Acta B. 2006;61(1):2–30.
  • Kieft IE, Broers JL, Caubet-Hilloutou V, et al. Electric discharge plasmas influence attachment of cultured CHO K1 cells. Bioelectromagnetics. 2004;25(5):362–368.
  • Guerrero-Preston R, Ogawa T, Uemura M, et al. Cold atmospheric plasma treatment selectively targets head and neck squamous cell carcinoma cells. Int J Mol Med. 2014;34(4):941–946.
  • Welz C, Emmert S, Canis M, et al. Cold atmospheric plasma: a promising complementary therapy for squamous head and neck cancer. PLoS One. 2015;10(11):e0141827.
  • Kim JY, Wei Y, Li J, et al. 15-Mum-sized single-cellular-level and cell-manipulatable microplasma jet in cancer therapies. Biosens Bioelectron. 2010;26(2):555–559.
  • Kaushik NK, Kaushik N, Park D, et al. Altered antioxidant system stimulates dielectric barrier discharge plasma-induced cell death for solid tumor cell treatment. PLoS One. 2014;9(7):e103349.
  • 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(12):e81576.
  • Hara H, Taniguchi M, Kobayashi M, et al. Plasma-activated medium-induced intracellular zinc liberation causes death of SH-SY5Y cells. Arch Biochem Biophys. 2015;584:51–60.
  • Tanaka H, Mizuno M, Toyokuni S, et al. Plasma-activated solution: mechanism of action, clinical application, and industrialization. Fukuoka Igaku Zasshi. 2015;106(4):71–76.
  • Ikeda JI, Tanaka H, Ishikawa K, et al. Plasma-activated medium (PAM) kills human cancer-initiating cells. Pathol Int. 2018;68(1):23–30.
  • Yoshikawa N, Liu W, Nakamura K, et al. Plasma-activated medium promotes autophagic cell death along with alteration of the mTOR pathway. Sci Rep. 2020;10(1):1614.
  • Hiromasa Tanaka MM, Ishikawa K, Nakamura K, et al. Cell survival and proliferation signaling pathways are downregulated by plasma-activated medium in glioblastoma brain tumor cells. Plasma Med. 2012;2(4):207–220.
  • Wende K, von Woedtke T, Weltmann KD, et al. Chemistry and biochemistry of cold physical plasma derived reactive species in liquids. Biol Chem. 2018;400(1):19–38.
  • 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;26:101291.
  • Nakamura K, Yoshikawa N, Mizuno Y, et al. Preclinical verification of the efficacy and safety of aqueous plasma for ovarian cancer therapy. Cancers. 2021;13(5):1141.
  • Tanaka H, Hori M. Medical applications of non-thermal atmospheric pressure plasma. J Clin Biochem Nutr. 2017;60(1):29–32.
  • Burm KTAL. Plasma: the fourth state of matter. Plasma Chem Plasma Process. 2012;32(2):401–407.
  • Nakamura K, Yoshikawa N, Yoshihara M, et al. Adjusted multiple gases in the plasma flow induce differential antitumor potentials of plasma-activated solutions. Plasma Process Polym. 2020;17(10):e1900259.
  • Cai Z, Yan LJ. Protein oxidative modifications: beneficial roles in disease and health. J Biochem Pharmacol Res. 2013;1(1):15–26.
  • Sies H. Strategies of antioxidant defense. Eur J Biochem. 1993;215(2):213–219.
  • Bauer G, Sersenová D, Graves DB, et al. Dynamics of singlet oxygen-triggered, RONS-based apoptosis induction after treatment of tumor cells with cold atmospheric plasma or plasma-activated medium. Sci Rep. 2019;9(1):13931.
  • Adachi T, Tanaka H, Nonomura S, et al. 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.
  • Tanaka H, Hosoi Y, Ishikawa K, et al. Low temperature plasma irradiation products of sodium lactate solution that induce cell death on U251SP glioblastoma cells were identified. Sci Rep. 2021;11(1):18488.
  • Zhen X, Sun HN, Liu R, et al. Non-thermal plasma-activated medium induces apoptosis of Aspc1 cells through the ROS-dependent autophagy pathway. In Vivo. 2020;34(1):143–153.
  • Jo A, Joh HM, Bae JH, et al. Plasma activated medium prepared by a bipolar microsecond-pulsed atmospheric pressure plasma jet array induces mitochondria-mediated apoptosis in human cervical cancer cells. PLoS One. 2022;17(8):e0272805.
  • Chauvin J, Gibot L, Griseti E, et al. Elucidation of in vitro cellular steps induced by antitumor treatment with plasma-activated medium. Sci Rep. 2019;9(1):4866.
  • Judée F, Fongia C, Ducommun B, et al. Short and long time effects of low temperature plasma activated media on 3D multicellular tumor spheroids. Sci Rep. 2016;6:21421.
  • Sato Y, Yamada S, Takeda S, et al. Effect of plasma-activated lactated Ringer’s solution on pancreatic cancer cells in vitro and in vivo. Ann Surg Oncol. 2018;25(1):299–307.
  • Zhang H, Zhang J, Guo B, et al. The antitumor effects of plasma-activated saline on muscle-invasive bladder cancer cells in vitro and in vivo demonstrate its feasibility as a potential therapeutic approach. Cancers. 2021;13(5):1042.
  • Hattori N, Yamada S, Torii K, et al. Effectiveness of plasma treatment on pancreatic cancer cells. Int J Oncol. 2015;47(5):1655–1662.
  • Kurake N, Tanaka H, Ishikawa K, et al. Cell survival of glioblastoma grown in medium containing hydrogen peroxide and/or nitrite, or in plasma-activated medium. Arch Biochem Biophys. 2016;605:102–108.
  • Adachi T, Nonomura S, Horiba M, et al. Iron stimulates plasma-activated medium-induced A549 cell injury. Sci Rep. 2016;6:20928.
  • Saito K, Asai T, Fujiwara K, et al. Tumor-selective mitochondrial network collapse induced by atmospheric gas plasma-activated medium. Oncotarget. 2016;7(15):19910–19927.
  • Utsumi F, Kajiyama H, Nakamura K, et al. Variable susceptibility of ovarian cancer cells to non-thermal plasma-activated medium. Oncol Rep. 2016;35(6):3169–3177.
  • Adachi T, Kano A, Nonomura S, et al. Histone deacetylase inhibitors stimulate the susceptibility of A549 cells to a plasma-activated medium treatment. Arch Biochem Biophys. 2016;606:120–127.
  • Tanaka H, Nakamura K, Mizuno M, et al. Non-thermal atmospheric pressure plasma activates lactate in Ringer’s solution for anti-tumor effects. Sci Rep. 2016;6:36282.
  • 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. 2017;24(5):1188–1194.
  • Hara H, Sueyoshi S, Taniguchi M, et al. Differences in intracellular mobile zinc levels affect susceptibility to plasma-activated medium-induced cytotoxicity. Free Radic Res. 2017;51(3):306–315.
  • Li Y, Ho Kang M, Sup Uhm H, et al. Effects of atmospheric-pressure non-thermal bio-compatible plasma and plasma activated nitric oxide water on cervical cancer cells. Sci Rep. 2017;7:45781.
  • Nakamura K, Peng Y, Utsumi F, et al. Novel intraperitoneal treatment with non-thermal plasma-activated medium inhibits metastatic potential of ovarian cancer cells. Sci Rep. 2017;7(1):6085.
  • Van Boxem W, Van der Paal J, Gorbanev Y, et al. Anti-cancer capacity of plasma-treated PBS: effect of chemical composition on cancer cell cytotoxicity. Sci Rep. 2017;7(1):16478.
  • Shi L, Yu L, Zou F, et al. 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.
  • Chauvin J, Judee F, Merbahi N, et al. Effects of plasma activated medium on head and neck FaDu cancerous cells: comparison of 3D and 2D response. Anticancer Agents Med Chem. 2018;18(6):776–783.
  • Xiang L, Xu X, Zhang S, et al. 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.
  • Matsuzaki T, Kano A, Kamiya T, et al. Enhanced ability of plasma-activated lactated Ringer’s solution to induce A549 cell injury. Arch Biochem Biophys. 2018;656:19–30.
  • Mokhtari H, Farahmand L, Yaserian K, et al. The antiproliferative effects of cold atmospheric plasma-activated media on different cancer cell lines, the implication of ozone as a possible underlying mechanism. J Cell Physiol. 2018;234(5):6778–6782.
  • Kurake N, Ishikawa K, Tanaka H, et al. Non-thermal plasma-activated medium modified metabolomic profiles in the glycolysis of U251SP glioblastoma. Arch Biochem Biophys. 2019;662:83–92.
  • Wang L, Yang X, Yang C, et al. The inhibition effect of cold atmospheric plasma-activated media in cutaneous squamous carcinoma cells. Future Oncol. 2019;15(5):495–505.
  • Azzariti A, Iacobazzi RM, Di Fonte R, et al. Plasma-activated medium triggers cell death and the presentation of immune activating danger signals in melanoma and pancreatic cancer cells. Sci Rep. 2019;9(1):4099.
  • Griseti E, Kolosnjaj-Tabi J, Gibot L, et al. Pulsed electric field treatment enhances the cytotoxicity of plasma-activated liquids in a three-dimensional human colorectal cancer cell model. Sci Rep. 2019;9(1):7583.
  • Tornin J, Mateu-Sanz M, Rodríguez A, et al. Pyruvate plays a main role in the antitumoral selectivity of cold atmospheric plasma in osteosarcoma. Sci Rep. 2019;9(1):10681.
  • Tanaka H, Mizuno M, Katsumata Y, et al. Oxidative stress-dependent and -independent death of glioblastoma cells induced by non-thermal plasma-exposed solutions. Sci Rep. 2019;9(1):13657.
  • Nagaya M, Hara H, Kamiya T, et al. Inhibition of NAMPT markedly enhances plasma-activated medium-induced cell death in human breast cancer MDA-MB-231 cells. Arch Biochem Biophys. 2019;676:108155.
  • Mateu-Sanz M, Tornín J, Brulin B, et al. Cold plasma-treated Ringer’s saline: a weapon to target osteosarcoma. Cancers. 2020;12(1):227.
  • Bisag A, Bucci C, Coluccelli S, et al. Plasma-activated Ringer’s lactate solution displays a selective cytotoxic effect on ovarian cancer cells. Cancers. 2020;12(2):476.
  • Griseti E, Merbahi N, Golzio M. Anti-cancer potential of two plasma-activated liquids: implication of long-lived reactive oxygen and nitrogen species. Cancers. 2020;12(3):721.
  • Yang X, Yang C, Wang L, et al. Inhibition of basal cell carcinoma cells by cold atmospheric plasma‑activated solution and differential gene expression analysis. Int J Oncol. 2020;56(5):1262–1273.
  • Xu S, Wang Y, Que Y, et al. Cold atmospheric plasma‑activated Ringer’s solution inhibits the proliferation of osteosarcoma cells through the mitochondrial apoptosis pathway. Oncol Rep. 2020;43(5):1683–1691.
  • Hwang SY, Nguyen NH, Kim TJ, et al. Non-thermal plasma couples oxidative stress to TRAIL sensitization through DR5 upregulation. Int J Mol Sci. 2020;21(15):5302.
  • Jo A, Joh HM, Chung TH, et al. Anticancer effects of plasma-activated medium produced by a microwave-excited atmospheric pressure argon plasma jet. Oxid Med Cell Longev. 2020;2020:4205640.
  • Ishii R, Kamiya T, Hara H, et al. Hyperthermia synergistically enhances cancer cell death by plasma-activated acetated Ringer’s solution. Arch Biochem Biophys. 2020;693:108565.
  • Cheng YJ, Lin CK, Chen CY, et al. Plasma-activated medium as adjuvant therapy for lung cancer with malignant pleural effusion. Sci Rep. 2020;10(1):18154.
  • Adachi T, Matsuda Y, Ishii R, et al. Ability of plasma-activated acetated Ringer’s solution to induce A549 cell injury is enhanced by a pre-treatment with histone deacetylase inhibitors. J Clin Biochem Nutr. 2020;67(3):232–239.
  • Tornín J, Villasante A, Solé-Martí X, et al. Osteosarcoma tissue-engineered model challenges oxidative stress therapy revealing promoted cancer stem cell properties. Free Radic Biol Med. 2021;164:107–118.
  • Sklias K, Santos Sousa J, Girard PM. Role of short- and long-lived reactive species on the selectivity and anti-cancer action of plasma treatment in vitro. Cancers. 2021;13(4):615.
  • Eto K, Ishinada C, Suemoto T, et al. Differential data on the responsiveness of multiple cell types to cell death induced by non-thermal atmospheric pressure plasma-activated solutions. Data Brief. 2021;36:106995.
  • Tomić S, Petrović A, Puač N, et al. Plasma-activated medium potentiates the immunogenicity of tumor cell lysates for dendritic cell-based cancer vaccines. Cancers. 2021;13(7):1626.
  • Tanaka H, Bekeschus S, Yan D, et al. Plasma-treated solutions (PTS) in cancer therapy. Cancers. 2021;13(7):1737.
  • Li Y, Tang T, Lee HJ, et al. Selective anti-cancer effects of plasma-activated medium and its high efficacy with cisplatin on hepatocellular carcinoma with cancer stem cell characteristics. Int J Mol Sci. 2021;22(8):3956
  • Jiang L, Zheng H, Lyu Q, et al. Lysosomal nitric oxide determines transition from autophagy to ferroptosis after exposure to plasma-activated Ringer’s lactate. Redox Biol. 2021;43:101989.
  • Silva-Teixeira R, Laranjo M, Lopes B, et al. Plasma activated media and direct exposition can selectively ablate retinoblastoma cells. Free Radic Biol Med. 2021;171:302–313.
  • Li Y, Tang T, Lee H, et al. Cold atmospheric pressure plasma-activated medium induces selective cell death in human hepatocellular carcinoma cells independently of singlet oxygen, hydrogen peroxide, nitric oxide and nitrite/nitrate. Int J Mol Sci. 2021;22(11):5548.
  • Ando T, Suzuki-Karasaki M, Suzuki-Karasaki M, et al. Combined anticancer effect of plasma-activated infusion and salinomycin by targeting autophagy and mitochondrial morphology. Front Oncol. 2021;11:593127.
  • Wang P, Zhou R, Thomas P, et al. Epithelial-to-mesenchymal transition enhances cancer cell sensitivity to cytotoxic effects of cold atmospheric plasmas in breast and bladder cancer systems. Cancers. 2021;13(12):2889.
  • Sersenová D, Machala Z, Repiská V, et al. Selective apoptotic effect of plasma activated liquids on human cancer cell lines. Molecules. 2021;26(14):4254.
  • Oh C, Won HR, Kang WS, et al. Head and neck cancer cell death due to mitochondrial damage induced by reactive oxygen species from nonthermal plasma-activated media: based on transcriptomic analysis. Oxid Med Cell Longev. 2021;2021:9951712.
  • Suzuki-Karasaki M, Ando T, Ochiai Y, et al. Air plasma-activated medium evokes a death-associated perinuclear mitochondrial clustering. Int J Mol Sci. 2022;23(3):1124.
  • Rebl H, Sawade M, Hein M, et al. Publisher correction: synergistic effect of plasma-activated medium and novel indirubin derivatives on human skin cancer cells by activation of the AhR pathway. Sci Rep. 2022;12(1):4341.
  • Jo A, Bae JH, Yoon YJ, et al. Plasma-activated medium induces ferroptosis by depleting FSP1 in human lung cancer cells. Cell Death Dis. 2022;13(3):212.
  • Lee YJ, Kim SW, Jung MH, et al. Plasma-activated medium inhibits cancer stem cell-like properties and exhibits a synergistic effect in combination with cisplatin in ovarian cancer. Free Radic Biol Med. 2022;182:276–288.
  • Zahedian S, Hekmat A, Tackallou SH, et al. The impacts of prepared plasma-activated medium (PAM) combined with doxorubicin on the viability of MCF-7 breast cancer cells: a new cancer treatment strategy. Rep Biochem Mol Biol. 2022;10(4):640–652.
  • Qi M, Xu D, Wang S, et al. In vivo metabolic analysis of the anticancer effects of plasma-activated saline in three tumor animal models. Biomedicines. 2022;10(3):528.
  • Li Y, Lv Y, Zhu Y, et al. Low-temperature plasma-activated medium inhibited proliferation and progression of lung cancer by targeting the PI3K/akt and MAPK pathways. Oxid Med Cell Longev. 2022;2022:9014501.
  • Sun HN, Guo XY, Xie DP, et al. Knockdown of peroxiredoxin V increased the cytotoxicity of non-thermal plasma-treated culture medium to A549 cells. Aging (Albany NY). 2022;14(9):4000–4013.
  • Mihai CT, Mihaila I, Pasare MA, et al. Cold atmospheric plasma-activated media improve paclitaxel efficacy on breast cancer cells in a combined treatment model. Curr Issues Mol Biol. 2022;44(5):1995–2014.
  • Gunes S, He Z, Tsoukou E, et al. Cell death induced in glioblastoma cells by plasma-activated-liquids (PAL) is primarily mediated by membrane lipid peroxidation and not ROS influx. PLoS One. 2022;17(9):e0274524.
  • Okazaki Y, Wang Y, Tanaka H, et al. Direct exposure of non-equilibrium atmospheric pressure plasma confers simultaneous oxidative and ultraviolet modifications in biomolecules. J Clin Biochem Nutr. 2014;55(3):207–215.
  • Gorlach A, Bertram K, Hudecova S, et al. Calcium and ROS: a mutual interplay. Redox Biol. 2015;6:260–271.
  • Schneider C, Gebhardt L, Arndt S, et al. Cold atmospheric plasma causes a calcium influx in melanoma cells triggering CAP-induced senescence. Sci Rep. 2018;8(1):10048.
  • Zheng G, Fu Y, He C. Nucleic acid oxidation in DNA damage repair and epigenetics. Chem Rev. 2014;114(8):4602–4620.
  • Ahn HJ, Kim KI, Kim G, et al. Atmospheric-pressure plasma jet induces apoptosis involving mitochondria via generation of free radicals. PLoS One. 2011;6(11):e28154.
  • Wada N, Ikeda JI, Tanaka H, et al. Effect of plasma-activated medium on the decrease of tumorigenic population in lymphoma. Pathol Res Pract. 2017;213(7):773–777.
  • Chang JW, Kang SU, Shin YS, et al. Non-thermal atmospheric pressure plasma induces apoptosis in oral cavity squamous cell carcinoma: involvement of DNA-damage-triggering Sub-G(1) arrest via the ATM/p53 pathway. Arch Biochem Biophys. 2014;545:133–140.
  • Ishaq M, Bazaka K, Ostrikov K. Intracellular effects of atmospheric-pressure plasmas on melanoma cancer cells. Phys Plasmas. 2015;22(12):122003.
  • Salimian Rizi B, Achreja A, Nagrath D. Nitric oxide: the forgotten child of tumor metabolism. Trends Cancer. 2017;3(9):659–672.
  • Girard PM, Arbabian A, Fleury M, et al. Synergistic effect of H2O2 and NO2 in cell death induced by cold atmospheric He plasma. Sci Rep. 2016;6:29098.
  • Moncada S, Erusalimsky JD. Does nitric oxide modulate mitochondrial energy generation and apoptosis? Nat Rev Mol Cell Biol. 2002;3(3):214–220.
  • Yan D, Talbot A, Nourmohammadi N, et al. Principles of using cold atmospheric plasma stimulated media for cancer treatment. Sci Rep. 2015;5:18339.
  • Baghba R, Roshangar L, Jahanban-Esfahlan R, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Communic Signal. 2020;18(1):59.
  • Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20(3):174–186.
  • Zheng AP, Wei YH, Zhao YN, et al. The role of cancer-associated mesothelial cells in the progression and therapy of ovarian cancer. Front Immunol. 2022;13:1013506.
  • Lin A, Truong B, Pappas 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 Process Polym. 2015;12(12):1392–1399.
  • Fucikova J, Kepp O, Kasikova L, et al. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11(11):1013.
  • Lin AG, Xiang B, Merlino DJ, et al. Non-thermal plasma induces immunogenic cell death in vivo in murine CT26 colorectal tumors. Oncoimmunology. 2018;7(9):e1484978.
  • Mizuno K, Yonetamari K, Shirakawa Y, et al. Anti-tumor immune response induced by nanosecond pulsed streamer discharge in mice. J Phys D Appl Phys. 2017;50(12):12LT01.
  • Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16(6):356–371.
  • Wang Y, Sun J, Zheng R, et al. Regulatory T cells are an important prognostic factor in breast cancer: a systematic review and meta-analysis. Neoplasma. 2016;63(5):789–798.
  • Shang B, Liu Y, Jiang SJ, et al. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep. 2015;5:15179.
  • Liedtke KR, Freund E, Hackbarth C, et al. A myeloid and lymphoid infiltrate in murine pancreatic tumors exposed to plasma-treated medium. Clin Plasma Med. 2018;11:10–17.
  • Zhou J, Tang Z, Gao S, et al. Tumor-associated macrophages: recent insights and therapies. Front Oncol. 2020;10:188.
  • Vayrynen JP, Haruki K, Lau MC, et al. The prognostic role of macrophage polarization in the colorectal cancer microenvironment. Cancer Immunol Res. 2021;9(1):8–19.
  • Lopez-Janeiro A, Padilla-Ansala C, de Andrea CE, et al. Prognostic value of macrophage polarization markers in epithelial neoplasms and melanoma. A systematic review and meta-analysis. Mod Pathol. 2020;33(8):1458–1465.
  • Kaushik NK, Kaushik N, Min B, et al. Cytotoxic macrophage-released tumour necrosis factor-alpha (TNF-alpha) as a killing mechanism for cancer cell death after cold plasma activation. J Phys D Appl Phys. 2016;49(8):084001.
  • Kaushik NK, Kaushik N, Adhikari M, et al. Preventing the solid cancer progression via release of anticancer-cytokines in co-culture with cold plasma-stimulated macrophages. Cancers. 2019;11(6):842.
  • Nastasa V, Pasca AS, Malancus RN, et al. Toxicity assessment of long-term exposure to non-thermal plasma activated water in mice. Int J Mol Sci. 2021;22(21):11534.
  • Xu DH, Cui QJ, Xu YJ, et al. Systemic study on the safety of immuno-deficient nude mice treated by atmospheric plasma-activated water. Plasma Sci Technol. 2018;20(4):044003. 10.1088/2058-6272/aa9842
  • Xu YJ, Peng SS, Li B, et al. Systematic safety evaluation of cold plasma-activated liquid in rabbits. Front Phys Lausanne. 2021;9:1700.

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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