199
Views
0
CrossRef citations to date
0
Altmetric
Preliminary Communication

Monitoring Nanoparticle-Mediated Cellular Hyperthermia with A High-Sensitivity Biosensor

, , , , , , , , , & show all
Pages 2729-2743 | Received 10 May 2013, Accepted 31 Oct 2013, Published online: 18 Feb 2014

References

  • Ahmed N , FessiH, ElaissariA. Theranostic applications of nanoparticles in cancer. Drug Discov. Today17, 928–934 (2012).
  • Chatterjee DK , DiagaradjaneP, KrishnanS. Nanoparticle-mediated hyperthermia in cancer therapy. Ther. Deliv.2(8), 1001–1014 (2011).
  • Nielsen OS , HorsmanM, OvergaardJ. A future for hyperthermia in cancer treatment? Eur. J. Cancer37(13), 1587–1589 (2001).
  • Gordon RT , HinesJR, GordonD. Intracellular hyperthermia. A biophysical approach to cancer treatment via intracellular temperature and biophysical alterations. Med. Hypotheses5(1), 83–102 (1979).
  • Basel MT , BalivadaS, WangHet al. Cell-delivered magnetic nanoparticles caused hyperthermia-mediated increased survival in a murine pancreatic cancer model. Int. J. Nanomedicine 7, 297–306 (2012).
  • Giustini AJ , IvkovR, HoopesPJ. Magnetic nanoparticle biodistribution following intratumoral administration. Nanotechnology22(34), 345101 (2011).
  • Johannsen M , GneveckowU, ThiesenBet al. Thermotherapy of prostate cancer using magnetic nanoparticles: feasibility, imaging, and three-dimensional temperature distribution. Eur. Urol. 52(6), 1653–1661 (2007).
  • Maier-Hauff K , UlrichF, NestlerDet al. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J. Neurooncol. 103(2), 317–324 (2011).
  • Dewey WC . Arrhenius relationships from the molecule and cell to the clinic. Int. J. Hyperthermia25(1), 3–20 (2009).
  • Kariya A , TabuchiY, YunokiT, KondoT. Identification of common gene networks responsive to mild hyperthermia in human cancer cells. Int. J. Mol. Med.32(1), 195–202 (2013).
  • Roti Roti JL . Cellular responses to hyperthermia (40–46°C): cell killing and molecular events. Int. J. Hyperthermia24(1), 3–15 (2008).
  • Krawczyk PM , EppinkB, EssersJet al. Mild hyperthermia inhibits homologous recombination, induces BRCA2 degradation, and sensitizes cancer cells to poly(ADP-ribose) polymerase-1 inhibition. Proc. Natl Acad. Sci. USA 108(24), 9851–9856 (2011).
  • Creixell M , BohorquezAC, Torres-LugoM, RinaldiC. EGFR-targeted magnetic nanoparticle heaters kill cancer cells without a perceptible temperature rise. ACS Nano5(9), 7124–7129 (2011).
  • Asin L , IbarraMR, TresA, GoyaGF. Controlled cell death by magnetic hyperthermia: effects of exposure time, field amplitude, and nanoparticle concentration. Pharm. Res.29(5), 1319–1327 (2012).
  • Villanueva A , De La Presa P, Alonso JM et al. Hyperthermia HeLa cell Treatment with silica-coated manganese oxide nanoparticles. J. Phys. Chem. C114(5), 1976–1981 (2010).
  • Krpetic Z , NativoP, SeeV, PriorIA, BrustM, VolkM. Inflicting controlled nonthermal damage to subcellular structures by laser-activated gold nanoparticles. Nano Lett.10(11), 4549–4554 (2010).
  • Jordan A , ScholzR, WustPet al. Endocytosis of dextran and silan-coated magnetite nanoparticles and the effect of intracellular hyperthermia on human mammary carcinoma cells in vitro. J. Magnet. Magn. Mater. 194 (1–3), 185–196 (1999).
  • Rodriguez-Luccioni HL , Latorre-EstevesM, Mendez-VegaJet al. Enhanced reduction in cell viability by hyperthermia induced by magnetic nanoparticles. Int. J. Nanomedicine 6, 373–380 (2011).
  • Gota C , OkabeK, FunatsuT, HaradaY, UchiyamaS. Hydrophilic fluorescent nanogel thermometer for intracellular thermometry. J. Am. Chem. Soc.131(8), 2766–2767 (2009).
  • Huang H , DelikanliS, ZengH, FerkeyDM, PralleA. Remote control of ion channels and neurons through magnetic-field heating of nanoparticles. Nat. Nanotechnol.5(8), 602–606 (2010).
  • Yang JM , YangH, LinL. Quantum dot nano thermometers reveal heterogeneous local thermogenesis in living cells. ACS Nano5(6), 5067–5071 (2011).
  • Raoof M , ZhuC, KaluarachchiWD, CurleySA. Luciferase-based protein denaturation assay for quantification of radiofrequency field-induced targeted hyperthermia: Developing an intracellular thermometer. Int. J. Hyperthermia28(3), 202–209 (2012).
  • Deckers R , DebeissatC, FortinPY, MoonenCT, CouillaudF. Arrhenius analysis of the relationship between hyperthermia and Hsp70 promoter activation: a comparison between ex vivo and in vivo data. Int. J. Hyperthermia28(5), 441–450 (2012).
  • Lupold SE , JohnsonT, ChowdhuryWH, RodriguezR. A real time Metridia luciferase based non-invasive reporter assay of mammalian cell viability and cytotoxicity via the beta-actin promoter and enhancer. PLoS ONE7(5), e36535 (2012).
  • Pelham HR . A regulatory upstream promoter element in the Drosophila hsp70 heat-shock gene. Cell30(2), 517–528 (1982).
  • Wu BJ , KingstonRE, MorimotoRI. Human HSP70 promoter contains at least two distinct regulatory domains. Proc. Natl Acad. Sci. USA83(3), 629–633 (1986).
  • Hedayati M , ThomasO, Abubaker-SharifBet al. The effect of cell-cluster size on intracellular nanoparticle-mediated hyperthermia: is it possible to treat microscopic tumors? Nanomedicine (Lond.) 8, 29–41 (2012).
  • Bordelon DE , CornejoC, GruettnerC, WestphalF, DeweeseTL, IvkovR. Magnetic nanoparticle heating efficiency reveals magneto-structural differences when characterized with wide ranging and high amplitude alternating magnetic fields. J. Appl. Phys.109(12), 124904 (2011).
  • Joinpoint Regression Program. http://surveillance.cancer.gov/joinpoint
  • Di YP , RepaskyEA, SubjeckJR. Distribution of HSP70, protein kinase C, and spectrin is altered in lymphocytes during a fever-like hyperthermia exposure. J. Cell Physiol.172(1), 44–54 (1997).
  • Park HG , HanSI, OhSY, KangHS. Cellular responses to mild heat stress. Cell. Mol. Life Sci.62(1), 10–23 (2005).
  • Kim HJ , FayMP, FeuerEJ, MidthuneDN. Permutation tests for joinpoint regression with applications to cancer rates. Stat. Med.19(3), 335–351 (2000).
  • Field SB , MorrisCC. The relationship between heating time and temperature: its relevance to clinical hyperthermia. Radiother. Oncol.1(2), 179–186 (1983).
  • Overgaard J , SuitHD. Time-temperature relationship th hyperthermic treatment of malignant and normal tissue in vivo. Cancer Res.39(8), 3248–3253 (1979).
  • Gruettner C , MuellerK, TellerJ, WestphalF, ForemanA, IvkovR. Synthesis and antibody conjugation of magnetic nanoparticles with improved specific power absorption rates for alternating magnetic field cancer therapy. J. Magnet. Magn. Mater.311(1), 181–186 (2007).
  • Klein S , SommerA, DistelLV, NeuhuberW, KryschiC. Superparamagnetic iron oxide nanoparticles as radiosensitizer via enhanced reactive oxygen species formation. Biochem. Biophys. Res. Commun.425(2), 393–397 (2012).
  • Voinov MA , Sosa Pagan JO, Morrison E, Smirnova TI, Smirnov AIi. Surface-mediated production of hydroxyl radicals as a mechanism of iron oxide nanoparticle biotoxicity. J. Am. Chem. Soc.133(1), 35–41 (2011).
  • Asin L , GoyaGF, TresA, IbarraMR. Induced cell toxicity originates dendritic cell death following magnetic hyperthermia treatment. Cell Death Dis.4, e596 (2013).

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.