519
Views
0
CrossRef citations to date
0
Altmetric
Perspective

Nanoparticle-mediated Hyperthermia in Cancer Therapy

, &
Pages 1001-1014 | Published online: 31 Aug 2011

Bibliography

  • Coley WB . II. Contribution to the Knowledge of Sarcoma. Ann. Surg.14(3), 199–220 (1891).
  • Friedenthal E , MendeckiJ, BotsteinC, SterzerF, NowogrodzkiM, PaglioneR. Some practical considerations for the use of localized hyperthermia in the treatment of cancer. J. Microw. Power16(2), 199–204 (1981).
  • Lele PP . Induction of deep, local hyperthermia by ultrasound and electromagnetic fields: problems and choices. Radiat. Environ. Biophys.17(3), 205–217 (1980).
  • Magin RL , JohnsonRK. Effects of local tumor hyperthermia on the growth of solid mouse tumors. Cancer Res.39(11), 4534–4539 (1979).
  • Kim JH , HahnEW, AhmedSA. Combination hyperthermia and radiation therapy for malignant melanoma. Cancer50(3), 478–482 (1982).
  • Luk KH , FrancisME, PerezCA, JohnsonRJ. Combined radiation and hyperthermia: comparison of two treatment schedules based on data from a registry established by the Radiation Therapy Oncology Group (RTOG). Int. J. Radiat. Oncol. Biol. Phys.10(6), 801–809 (1984).
  • Stewart JR , GibbsFA Jr. Hyperthermia in the treatment of cancer. Perspectives on its promise and its problems. Cancer54(11 Suppl.), 2823–2830 (1984).
  • Thrall DE . Clinical requirements for localized hyperthermia in the patient. Radiat. Environ. Biophys.17(3), 229–232 (1980).
  • Doss JD , MccabeCW. A technique for localized heating in tissue: an adjunct to tumor therapy. Med. Instrum.10(1), 16–21 (1976).
  • Irish CE , BrownJ, GalenWPet al. Thermoradiotherapy for persistent cancer in previously irradiated fields. Cancer 57(12), 2275–2279 (1986).
  • Seegenschmiedt MH , SauerR, MiyamotoC, ChalalJA, BradyLW. Clinical experience with interstitial thermoradiotherapy for localized implantable pelvic tumors. Am. J. Clin. Oncol.16(3), 210–222 (1993).
  • Bass H , MooreJL, CoakleyWT. Lethality in mammalian cells due to hyperthermia under oxic and hypoxic conditions. Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med.33(1), 57–67 (1978).
  • Storm FK . Clinical hyperthermia and chemotherapy. Radiol. Clin. North Am.27(3), 621–627 (1989).
  • Urano M , KurodaM, NishimuraY. For the clinical application of thermochemotherapy given at mild temperatures. Int. J. Hyperthermia15(2), 79–107 (1999).
  • Shakil A , OsbornJL, SongCW. Changes in oxygenation status and blood flow in a rat tumor model by mild temperature hyperthermia. Int. J. Radiat. Oncol. Biol. Phys.43(4), 859–865 (1999).
  • Johannsen M , ThiesenB, WustP, JordanA. Magnetic nanoparticle hyperthermia for prostate cancer. Int. J. Hyperthermia26(8), 790–795 (2010).
  • Hurwitz MD , HansenJL, Prokopios-DavosSet al. Hyperthermia combined with radiation for the treatment of locally advanced prostate cancer: long-term results from Dana-Farber Cancer Institute study 94–153. Cancer 117(3), 510–516 (2011).
  • Moros EG , PenagaricanoJ, NovakP, StraubeWL, MyersonRJ. Present and future technology for simultaneous superficial thermoradiotherapy of breast cancer. Int. J. Hyperthermia26(7), 699–709 (2010).
  • Van Den Berg CA , Van De Kamer JB, De Leeuw AA et al. Towards patient specific thermal modelling of the prostate. Phys. Med. Biol.51(4), 809–825 (2006).
  • Zagar TM , OlesonJR, VujaskovicZet al. Hyperthermia combined with radiation therapy for superficial breast cancer and chest wall recurrence: a review of the randomised data. Int. J. Hyperthermia 26(7), 612–617 (2010).
  • Franckena M , Van Der Zee J. Use of combined radiation and hyperthermia for gynecological cancer. Curr. Opin. Obstet. Gynecol.22(1), 9–14 (2010).
  • Vasanthan A , MitsumoriM, ParkJHet al. Regional hyperthermia combined with radiotherapy for uterine cervical cancers: a multi-institutional prospective randomized trial of the international atomic energy agency. Int. J. Radiat. Oncol. Biol. Phys. 61(1), 145–153 (2005).
  • Huilgol NG , GuptaS, SridharCR. Hyperthermia with radiation in the treatment of locally advanced head and neck cancer: a report of randomized trial. J. Cancer Res. Ther.6(4), 492–496 (2010).
  • Huilgol NG , GuptaS, DixitR. Chemoradiation with hyperthermia in the treatment of head and neck cancer. Int. J. Hyperthermia26(1), 21–25 (2010).
  • Chang E , AlexanderHR, LibuttiSKet al. Laparoscopic continuous hyperthermic peritoneal perfusion. J. Am. Coll. Surg. 193(2), 225–229 (2001).
  • Feldman AL , LibuttiSK, PingpankJFet al. Analysis of factors associated with outcome in patients with malignant peritoneal mesothelioma undergoing surgical debulking and intraperitoneal chemotherapy. J. Clin. Oncol. 21(24), 4560–4567 (2003).
  • Maeda H . The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Adv. Enzyme Regul.41, 189–207 (2001).
  • Maeda H , WuJ, SawaT, MatsumuraY, HoriK. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J. Control. Release65(1–2), 271–284 (2000).
  • Kah JC , WongKY, NeohKGet al. Critical parameters in the pegylation of gold nanoshells for biomedical applications: an in vitro macrophage study. J. Drug Target 17(3), 181–193 (2009).
  • Wang J , SuiM, FanW. Nanoparticles for tumor targeted therapies and their pharmacokinetics. Curr. Drug Metab.11(2), 129–141 (2010).
  • El-Sayed IH , HuangX, El-SayedMA. Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett.239(1), 129–135 (2006).
  • Waldman SA , FortinaP, SurreyS, HyslopT, KrickaLJ, GravesDJ. Opportunities for near-infrared thermal ablation of colorectal metastases by guanylyl cyclase C-targeted gold nanoshells. Future Oncol.2(6), 705–716 (2006).
  • Patra CR , BhattacharyaR, MukhopadhyayD, MukherjeeP. Fabrication of gold nanoparticles for targeted therapy in pancreatic cancer. Adv. Drug Deliv. Rev.62(3), 346–361 (2010).
  • Hosta-Rigau L , OlmedoI, ArbiolJ, CruzLJ, KoganMJ, AlbericioF. Multifunctionalized gold nanoparticles with peptides targeted to gastrin-releasing peptide receptor of a tumor cell line. Bioconjug. Chem.21(6), 1070–1078 (2010).
  • Diagaradjane P , ShettyA, WangJCet al. Modulation of in vivo tumor radiation response via gold nanoshell-mediated vascular-focused hyperthermia: characterizing an integrated antihypoxic and localized vascular disrupting targeting strategy. Nano Lett. 8(5), 1492–1500 (2008).
  • Hirsch LR , GobinAM, LoweryARet al. Metal nanoshells. Ann. Biomed. Eng. 34(1), 15–22 (2006).
  • Hulander M , HongJ, AnderssonMet al. Blood interactions with noble metals: coagulation and immune complement activation. ACS Appl. Mater. Interfaces 1(5), 1053–1062 (2009).
  • Choi WI , KimJY, KangC, ByeonCC, KimYH, TaeG. Tumor regression in vivo by photothermal therapy based on gold-nanorod-loaded, functional nanocarriers. ACS Nano5(3), 1995–2003 (2011).
  • Von Maltzahn G , ParkJH, AgrawalAet al. Computationally guided photothermal tumor therapy using long-circulating gold nanorod antennas. Cancer Res. 69(9), 3892–3900 (2009).
  • Hirsch LR , StaffordRJ, BanksonJAet al. Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance. Proc. Natl Acad. Sci. USA 100(23), 13549–13554 (2003).
  • O‘neal DP , HirschLR, HalasNJ, PayneJD, WestJL. Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. Cancer Lett.209(2), 171–176 (2004).
  • Bernardi RJ , LoweryAR, ThompsonPA, BlaneySM, WestJL. Immunonanoshells for targeted photothermal ablation in medulloblastoma and glioma: an in vitro evaluation using human cell lines. J. Neurooncol.86(2), 165–172 (2008).
  • Cheng FY , ChenCT, YehCS. Comparative efficiencies of photothermal destruction of malignant cells using antibody-coated silica–Au nanoshells, hollow Au/Ag nanospheres and Au nanorods. Nanotechnology20(42), 425104 (2009).
  • Dickerson EB , DreadenEC, HuangXet al. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett. 269(1), 57–66 (2008).
  • Skrabalak SE , AuL, LuX, LiX, XiaY. Gold nanocages for cancer detection and treatment. Nanomedicine (Lond.)2(5), 657–668 (2007).
  • Atkinson RL , ZhangM, DiagaradjanePet al. Thermal enhancement with optically activated gold nanoshells sensitizes breast cancer stem cells to radiation therapy. Sci. Transl. Med. 2(55), 55ra79 (2010).
  • Gilchrist RK , MedalR, ShoreyWD, HanselmanRC, ParrottJC, TaylorCB. Selective inductive heating of lymph nodes. Ann. Surg.146(4), 596–606 (1957).
  • Yallapu MM , OthmanSF, CurtisET, GuptaBK, JaggiM, ChauhanSC. Multi-functional magnetic nanoparticles for magnetic resonance imaging and cancer therapy. Biomaterials32(7), 1890–1905 (2011).
  • Huang N , WangH, ZhaoJ, LuiH, KorbelikM, ZengH. Single-wall carbon nanotubes assisted photothermal cancer therapy: animal study with a murine model of squamous cell carcinoma. Lasers Surg. Med.42(9), 638–648
  • Elsherbini AA , SaberM, AggagM, El-ShahawyA, ShokierHA. Magnetic nanoparticle-induced hyperthermia treatment under magnetic resonance imaging. Magn. Reson. Imaging29(2), 272–280 (2011).
  • Majumdar S , ZoghbiSS, GoreJC. Pharmacokinetics of superparamagnetic iron-oxide MR contrast agents in the rat. Invest. Radiol.25(7), 771–777 (1990).
  • Okon E , PouliquenD, OkonPet al. Biodegradation of magnetite dextran nanoparticles in the rat. A histologic and biophysical study. Lab. Invest. 71(6), 895–903 (1994).
  • Wu X , TanY, MaoH, ZhangM. Toxic effects of iron oxide nanoparticles on human umbilical vein endothelial cells. Int. J. Nanomedicine5, 385–399 (2010).
  • Fan C , GaoW, ChenZet al. Tumor selectivity of stealth multi-functionalized superparamagnetic iron oxide nanoparticles. Int. J. Pharm. 404(1–2), 180–190 (2011).
  • Balivada S , RachakatlaRS, WangHet al. A/C magnetic hyperthermia of melanoma mediated by iron(0)/iron oxide core/shell magnetic nanoparticles: a mouse study. BMC Cancer 10, 119 (2010).
  • Rachakatla RS , BalivadaS, SeoGMet al. Attenuation of mouse melanoma by A/C magnetic field after delivery of bi-magnetic nanoparticles by neural progenitor cells. ACS Nano 4(12), 7093–7104 (2010).
  • Le Renard PE , JordanO, FaesAet al. The in vivo performance of magnetic particle-loaded injectable, in situ gelling, carriers for the delivery of local hyperthermia. Biomaterials 31(4), 691–705 (2010).
  • Hayashi K , OnoK, SuzukiHet al. High-frequency, magnetic-field-responsive drug release from magnetic nanoparticle/organic hybrid based on hyperthermic effect. ACS Appl. Mater. Interfaces 2(7), 1903–1911 (2010).
  • Mohammad F , BalajiG, WeberA, UppuRM, KumarCS. Influence of gold nanoshell on hyperthermia of superparamagnetic iron oxide nanoparticles (SPIONs). J. Phys. Chem. C Nanomater. Interfaces1(20), 3141–3146 (2010).
  • Zheng LX , O‘connellMJ, DoornSKet al. Ultralong single-wall carbon nanotubes. Nat. Mater. 3(10), 673–676 (2004).
  • Kam NW , O‘ConnellM, WisdomJA, DaiH. Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. Proc. Natl Acad. Sci. USA102(33), 11600–11605 (2005).
  • Biris AS , BoldorD, PalmerJet al. Nanophotothermolysis of multiple scattered cancer cells with carbon nanotubes guided by time-resolved infrared thermal imaging. J. Biomed. Opt. 14(2), 021007 (2009).
  • Torti SV , ByrneF, WhelanOet al. Thermal ablation therapeutics based on CN(x) multi-walled nanotubes. Int. J. Nanomedicine 2(4), 707–714 (2007).
  • Moon HK , LeeSH, ChoiHC. In vivo near-infrared mediated tumor destruction by photothermal effect of carbon nanotubes. ACS Nano3(11), 3707–3713 (2009).
  • Gannon CJ , CherukuriP, YakobsonBIet al. Carbon nanotube-enhanced thermal destruction of cancer cells in a noninvasive radiofrequency field. Cancer 110(12), 2654–2665 (2007).
  • Burke A , DingX, SinghRet al. Long-term survival following a single treatment of kidney tumors with multiwalled carbon nanotubes and near-infrared radiation. Proc. Natl Acad. Sci. USA 106(31), 12897–12902 (2009).
  • Poland CA , DuffinR, KinlochIet al. Carbon nanotubes introduced into the abdominal cavity of mice show asbestos-like pathogenicity in a pilot study. Nat. Nanotechnol. 3(7), 423–428 (2008).
  • Glazer ES , CurleySA. Radiofrequency field-induced thermal cytotoxicity in cancer cells treated with fluorescent nanoparticles. Cancer116(13), 3285–3293 (2010).
  • Melnikov OV , GorbenkoOY, MarkelovaMNet al. Ag-doped manganite nanoparticles: new materials for temperature-controlled medical hyperthermia. J. Biomed. Mater. Res. A 91(4), 1048–1055 (2009).
  • Fuller KJ , IsselsRD, SlosmanDO, GuilletJG, SoussiT, PollaBS. Cancer and the heat-shock response. Eur. J. Cancer30A(12), 1884–1891 (1994).
  • Harmon BV , TakanoYS, WinterfordCM, GobeGC. The role of apoptosis in the response of cells and tumours to mild hyperthermia. Int. J. Radiat. Biol.59(2), 489–501 (1991).

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.