1,309
Views
106
CrossRef citations to date
0
Altmetric
Review

Gold Nanoparticles as Radiosensitizers in Cancer Radiotherapy

, , ORCID Icon &
Pages 9407-9430 | Published online: 24 Nov 2020

References

  • Song G, Cheng L, Chao Y, Yang K, Liu Z. Emerging nanotechnology and advanced materials for cancer radiation therapy. Adv Mater. 2017;29(32):1700996.
  • Haume K, Rosa S, Grellet S, et al. Gold nanoparticles for cancer radiotherapy: a review. Cancer Nanotechnol. 2016;7(1):8.27867425
  • Her S, Jaffray DA, Allen C. Gold nanoparticles for applications in cancer radiotherapy: mechanisms and recent advancements. Adv Drug Deliv Rev. 2017;109:84–101. doi:10.1016/j.addr.2015.12.01226712711
  • Liu Y, Zhang P, Li F, et al. Metal-based nanoenhancers for future radiotherapy: radiosensitizing and synergistic effects on tumor cells. Theranostics. 2018;8(7):1824–1849. doi:10.7150/thno.2217229556359
  • Laprise-Pelletier M, Simão T, Fortin MA. Gold nanoparticles in radiotherapy and recent progress in nanobrachytherapy. Adv Healthc Mater. 2018;7(16):e1701460.29726118
  • Brun E, Sanche L, Sicard-Roselli C. Parameters governing gold nanoparticle X-ray radiosensitization of DNA in solution. Colloids Surf B Biointerfaces. 2009;72(1):128–134. doi:10.1016/j.colsurfb.2009.03.02519414242
  • Baumann M, Krause M, Overgaard J, et al. Radiation oncology in the era of precision medicine. Nat Rev Cancer. 2016;16(4):234–249.27009394
  • Nagi NMS, Khair YAM, Abdalla AME. Capacity of gold nanoparticles in cancer radiotherapy. Jpn J Radiol. 2017;35(10):555–561.28795273
  • Ngwa W, Kumar R, Sridhar S, et al. Targeted radiotherapy with gold nanoparticles: current status and future perspectives. Nanomedicine (Lond). 2014;9(7):1063–1082. doi:10.2217/nnm.14.5524978464
  • Ishikura S. Optimal radiotherapy for non-small-cell lung cancer: current progress and future challenges. Gen Thorac Cardiovasc Surg. 2012;60(3):127–131. doi:10.1007/s11748-011-0832-y22419179
  • Shrestha S, Cooper LN, Andreev OA, Reshetnyak YK, Antosh MP. Gold nanoparticles for radiation enhancement in vivo. Jacobs J Radiat Oncol. 2016;3(1):026.28725881
  • Linam J, Yang LX. Recent developments in radiosensitization. Anticancer Res. 2015;35(5):2479–2485.25964520
  • Mesbahi A. A review on gold nanoparticles radiosensitization effect in radiation therapy of cancer. Rep Pract Oncol Radiother. 2010;15(6):176–180. doi:10.1016/j.rpor.2010.09.00124376946
  • Guo Z, Zhu S, Yong Y, et al. Synthesis of BSA-coated BiOI@Bi S semiconductor heterojunction nanoparticles and their applications for radio/photodynamic/photothermal synergistic therapy of tumor. Adv Mater. 2017;29(44):1704136. doi:10.1002/adma.201704136
  • Ma N, Jiang YW, Zhang X, et al. Enhanced radiosensitization of gold nanospikes via hyperthermia in combined cancer radiation and photothermal therapy. ACS Appl Mater Interfaces. 2016;8(42):28480–28494. doi:10.1021/acsami.6b1013227689441
  • Zhang XD, Wu D, Shen X, et al. Size-dependent radiosensitization of PEG-coated gold nanoparticles for cancer radiation therapy. Biomaterials. 2012;33(27):6408–6419. doi:10.1016/j.biomaterials.2012.05.04722681980
  • Kobayashi K, Usami N, Porcel E, Lacombe S, Le Sech C. Enhancement of radiation effect by heavy elements. Mutat Res. 2010;704(1–3):123–131. doi:10.1016/j.mrrev.2010.01.00220074660
  • Yong Y, Cheng X, Bao T, et al. Tungsten sulfide quantum dots as multifunctional nanotheranostics for in vivo dual-modal image-guided photothermal/radiotherapy synergistic therapy. ACS Nano. 2015;9(12):12451–12463. doi:10.1021/acsnano.5b0582526495962
  • Wen L, Chen L, Zheng S, et al. Ultrasmall biocompatible WO3- x nanodots for multi-modality imaging and combined therapy of cancers. Adv Mater. 2016;28(25):5072–5079. doi:10.1002/adma.20150642827136070
  • Song G, Ji C, Liang C, et al. TaOx decorated perfluorocarbon nanodroplets as oxygen reservoirs to overcome tumor hypoxia and enhance cancer radiotherapy. Biomaterials. 2017;112:257–263. doi:10.1016/j.biomaterials.2016.10.02027768978
  • Liu J, Yang Y, Zhu W, et al. Nanoscale metal-organic frameworks for combined photodynamic & radiation therapy in cancer treatment. Biomaterials. 2016;97:1–9. doi:10.1016/j.biomaterials.2016.04.03427155362
  • Wu H, Lin J, Liu P, et al. Is the autophagy a friend or foe in the silver nanoparticles associated radiotherapy for glioma? Biomaterials. 2015;62:47–57. doi:10.1016/j.biomaterials.2015.05.03326022979
  • Kim SR, Kim EH. Feasibility study on the use of gold nanoparticles in fractionated kilovoltage X-ray treatment of melanoma. Int J Radiat Biol. 2018;94(1):8–16. doi:10.1080/09553002.2018.139357929034758
  • Dou Y, Guo Y, Li X, et al. Size-tuning ionization to optimize gold nanoparticles for simultaneous enhanced CT imaging and radiotherapy. ACS Nano. 2016;10(2):2536–2548. doi:10.1021/acsnano.5b0747326815933
  • Chang MY, Shiau AL, Chen YH, Chang CJ, Chen HH, Wu CL. Increased apoptotic potential and dose-enhancing effect of gold nanoparticles in combination with single-dose clinical electron beams on tumor-bearing mice. Cancer Sci. 2008;99(7):1479–1484. doi:10.1111/j.1349-7006.2008.00827.x18410403
  • Singh P, Pandit S, Mokkapati VRSS, Garg A, Ravikumar V, Mijakovic I. Gold nanoparticles in diagnostics and therapeutics for human cancer. Int J Mol Sci. 2018;19(7):1979. doi:10.3390/ijms19071979
  • Yang YS, Carney RP, Stellacci F, Irvine DJ. Enhancing radiotherapy by lipid nanocapsule-mediated delivery of amphiphilic gold nanoparticles to intracellular membranes. ACS Nano. 2014;8(9):8992–9002. doi:10.1021/nn502146r25123510
  • Kuncic Z, Lacombe S. Nanoparticle radio-enhancement: principles, progress and application to cancer treatment. Phys Med Biol. 2018;63(2):02TR01. doi:10.1088/1361-6560/aa99ce
  • Silva F, Paulo A, Pallier A, et al. Dual imaging gold nanoplatforms for targeted radiotheranostics. Materials (Basel). 2020;13(3):513. doi:10.3390/ma13030513
  • Siddique S, Chow JCL. Application of nanomaterials in biomedical imaging and cancer therapy. Nanomaterials (Basel). 2020;10(9):E1700.32872399
  • Yeh YC, Creran B, Rotello VM. Gold nanoparticles: preparation, properties, and applications in bionanotechnology. Nanoscale. 2012;4(6):1871–1880. doi:10.1039/C1NR11188D22076024
  • Alkilany AM, Thompson LB, Boulos SP, Sisco PN, Murphy CJ. Gold nanorods: their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Adv Drug Deliv Rev. 2012;64(2):190–199.21397647
  • Mieszawska AJ, Mulder WJ, Fayad ZA, Cormode DP. Multifunctional gold nanoparticles for diagnosis and therapy of disease. Mol Pharm. 2013;10(3):831–847. doi:10.1021/mp300588523360440
  • Rana S, Bajaj A, Mout R, Rotello VM. Monolayer coated gold nanoparticles for delivery applications. Adv Drug Deliv Rev. 2012;64(2):200–216. doi:10.1016/j.addr.2011.08.00621925556
  • Arvizo R, Bhattacharya R, Mukherjee P. Gold nanoparticles: opportunities and challenges in nanomedicine. Expert Opin Drug Deliv. 2010;7(6):753–763. doi:10.1517/1742524100377701020408736
  • Zheng T, Bott S, Huo Q. Techniques for accurate sizing of gold nanoparticles using dynamic light scattering with particular application to chemical and biological sensing based on aggregate formation. ACS Appl Mater Interfaces. 2016;8(33):21585–21594. doi:10.1021/acsami.6b0690327472008
  • Yu Y, Yang T, Sun T. New insights into the synthesis, toxicity and applications of gold nanoparticles in CT imaging and treatment of cancer. Nanomedicine (Lond). 2020;15(11):1127–1145. doi:10.2217/nnm-2019-039532329396
  • Guo J, Rahme K, He Y, Li LL, Holmes JD, O’Driscoll CM. Gold nanoparticles enlighten the future of cancer theranostics. Int J Nanomedicine. 2017;12:6131–6152. doi:10.2147/IJN.S14077228883725
  • Siddique S, Chow JCL. Gold nanoparticles for drug delivery and cancer therapy. Appl Sci. 2020;10(11):3824. doi:10.3390/app10113824
  • Babaei M, Ganjalikhani M. A systematic review of gold nanoparticles as novel cancer therapeutics. Nanomed J. 2014;1(4):211–219.
  • Huang K, Ma H, Liu J, et al. Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS Nano. 2012;6(5):4483–4493. doi:10.1021/nn301282m22540892
  • Ma N, Wu FG, Zhang X, et al. Shape-dependent radiosensitization effect of gold nanostructures in cancer radiotherapy: comparison of gold nanoparticles, nanospikes, and nanorods. ACS Appl Mater Interfaces. 2017;9(15):13037–13048.28338323
  • Li J, Li JJ, Zhang J, Wang X, Kawazoe N, Chen G. Gold nanoparticle size and shape influence on osteogenesis of mesenchymal stem cells. Nanoscale. 2016;8(15):7992–8007. doi:10.1039/C5NR08808A27010117
  • Kumar R, Korideck H, Ngwa W, Berbeco RI, Makrigiorgos GM, Sridhar S. Third generation gold nanoplatform optimized for radiation therapy. Transl Cancer Res. 2013;2(4). doi:10.3978/j.issn.2218-676X.2013.07.02
  • Caldorera-Moore M, Guimard N, Shi L, Roy K. Designer nanoparticles: incorporating size, shape and triggered release into nanoscale drug carriers. Expert Opin Drug Deliv. 2010;7(4):479–495. doi:10.1517/1742524090357997120331355
  • Morozov KV, Kolyvanova MA, Kartseva ME, et al. Radiosensitization by gold nanoparticles: impact of the size, dose rate, and photon energy. Nanomaterials (Basel). 2020;10(5):952. doi:10.3390/nano10050952
  • Jain S, Coulter JA, Butterworth KT, et al. Gold nanoparticle cellular uptake, toxicity and radiosensitisation in hypoxic conditions. Radiother Oncol. 2014;110(2):342–347. doi:10.1016/j.radonc.2013.12.01324444528
  • Gilles M, Brun E, Sicard-Roselli C. Gold nanoparticles functionalization notably decreases radiosensitization through hydroxyl radical production under ionizing radiation. Colloids Surf B Biointerfaces. 2014;123:770–777. doi:10.1016/j.colsurfb.2014.10.02825454667
  • Schuemann J, Berbeco R, Chithrani DB, et al. Roadmap to clinical use of gold nanoparticles for radiation sensitization. Int J Radiat Oncol Biol Phys. 2016;94(1):189–205. doi:10.1016/j.ijrobp.2015.09.03226700713
  • Coulter JA, Jain S, Butterworth KT, et al. Cell type-dependent uptake, localization, and cytotoxicity of 1.9 nm gold nanoparticles. Int J Nanomed. 2012;7:2673–2685. doi:10.2147/IJN.S31751
  • Hainfeld JF, Slatkin DN, Smilowitz HM. The use of gold nanoparticles to enhance radiotherapy in mice. Phys Med Biol. 2004;49(18):N309–N315. doi:10.1088/0031-9155/49/18/N0315509078
  • Hainfeld JF, Dilmanian FA, Zhong Z, Slatkin DN, Kalef-Ezra JA, Smilowitz HM. Gold nanoparticles enhance the radiation therapy of a murine squamous cell carcinoma. Phys Med Biol. 2010;55(11):3045–3059.20463371
  • Hainfeld JF, Smilowitz HM, O’Connor MJ, Dilmanian FA, Slatkin DN. Gold nanoparticle imaging and radiotherapy of brain tumors in mice. Nanomedicine (Lond). 2013;8(10):1601–1609. doi:10.2217/nnm.12.16523265347
  • Joh DY, Sun L, Stangl M, et al. Selective targeting of brain tumors with gold nanoparticle-induced radiosensitization. PLoS One. 2013;8(4):e62425. doi:10.1371/journal.pone.006242523638079
  • Cifter G, Chin J, Cifter F, et al. Targeted radiotherapy enhancement during electronic brachytherapy of accelerated partial breast irradiation (APBI) using controlled release of gold nanoparticles. Phys Med. 2015;31(8):1070–1074.26404139
  • Mousavie Anijdan SH, Mahdavi SR, Shirazi A, Zarrinfard MA, Hajati J. Megavoltage X-ray dose enhancement with gold nanoparticles in tumor bearing mice. Int J Mol Cell Med. 2013;2(3):118–123.24551801
  • Hau H, Khanal D, Rogers L, et al. Dose enhancement and cytotoxicity of gold nanoparticles in colon cancer cells when irradiated with kilo- and mega-voltage radiation. Bioeng Transl Med. 2016;1(1):94–102. doi:10.1002/btm2.1000729313009
  • Abbasian M, Baharlouei A, Arab-Bafrani Z, Lightfoot DA. Combination of gold nanoparticles with low-LET irradiation: an approach to enhance DNA DSB induction in HT29 colorectal cancer stem-like cells. J Cancer Res Clin Oncol. 2019;145(1):97–107. doi:10.1007/s00432-018-2769-330341685
  • Liu Y, Liu X, Jin X, et al. The dependence of radiation enhancement effect on the concentration of gold nanoparticles exposed to low- and high-LET radiations. Phys Med. 2015;31(3):210–218. doi:10.1016/j.ejmp.2015.01.00625651760
  • Kanavi MR, Asadi S, Balagholi S, Alikarami F, Nosrati H, Ahmadieh H. Gamma irradiation of ocular melanoma and lymphoma cells in the presence of gold nanoparticles: in vitro study. J Appl Clin Med Phys. 2018;19(3):268–275. doi:10.1002/acm2.1233629707895
  • Jain S, Hirst DG, O’Sullivan JM. Gold nanoparticles as novel agents for cancer therapy. Br J Radiol. 2012;85(1010):101–113. doi:10.1259/bjr/5944883322010024
  • Perrault SD, Walkey C, Jennings T, Fischer HC, Chan WC. Mediating tumor targeting efficiency of nanoparticles through design. Nano Lett. 2009;9(5):1909–1915. doi:10.1021/nl900031y19344179
  • Lechtman E, Chattopadhyay N, Cai Z, Mashouf S, Reilly R, Pignol JP. Implications on clinical scenario of gold nanoparticle radiosensitization in regards to photon energy, nanoparticle size, concentration and location. Phys Med Biol. 2011;56(15):4631–4647. doi:10.1088/0031-9155/56/15/00121734337
  • Chithrani DB, Jelveh S, Jalali F, et al. Gold nanoparticles as radiation sensitizers in cancer therapy. Radiat Res. 2010;173(6):719–728.20518651
  • Chithrani DB. Nanoparticles for improved therapeutics and imaging in cancer therapy. Recent Pat Nanotechnol. 2010;4(3):171–180. doi:10.2174/18722101079248372620618149
  • Wong C, Stylianopoulos T, Cui J, et al. Multistage nanoparticle delivery system for deep penetration into tumor tissue. Proc Natl Acad Sci U S A. 2011;108(6):2426–2431. doi:10.1073/pnas.101838210821245339
  • Cedervall T, Lynch I, Lindman S, et al. Understanding the nanoparticle-protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles. Proc Natl Acad Sci U S A. 2007;104(7):2050–2055. doi:10.1073/pnas.060858210417267609
  • Choi CH, Alabi CA, Webster P, Davis ME. Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles. Proc Natl Acad Sci U S A. 2010;107(3):1235–1240.20080552
  • Yasui H, Takeuchi R, Nagane M, et al. Radiosensitization of tumor cells through endoplasmic reticulum stress induced by PEGylated nanogel containing gold nanoparticles. Cancer Lett. 2014;347(1):151–158.24530512
  • Alalaiwe A, Roberts G, Carpinone P, Munson J, Roberts S. Influence of PEG coating on the oral bioavailability of gold nanoparticles in rats. Drug Deliv. 2017;24(1):591–598. doi:10.1080/10717544.2017.128255428222611
  • Hinkley GK, Carpinone P, Munson JW, Powers KW, Roberts SM. Oral absorption of PEG-coated versus uncoated gold nanospheres: does agglomeration matter? Part Fibre Toxicol. 2015;12:9. doi:10.1186/s12989-015-0085-525884802
  • Cao-Milán R, Liz-Marzán LM. Gold nanoparticle conjugates: recent advances toward clinical applications. Expert Opin Drug Deliv. 2014;11(5):741–752.24559075
  • Charbgoo F, Nejabat M, Abnous K, et al. Gold nanoparticle should understand protein corona for being a clinical nanomaterial. J Control Release. 2018;272:39–53. doi:10.1016/j.jconrel.2018.01.00229305922
  • Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. Nanoparticle PEGylation for imaging and therapy. Nanomedicine (Lond). 2011;6(4):715–728. doi:10.2217/nnm.11.1921718180
  • Liu S, Lämmerhofer M. Functionalized gold nanoparticles for sample preparation: a review. Electrophoresis. 2019;40:2438–2461.31056767
  • Mousavi M, Nedaei HA, Khoei S, et al. Enhancement of radiosensitivity of melanoma cells by pegylated gold nanoparticles under irradiation of megavoltage electrons. Int J Radiat Biol. 2017;93(2):214–221. doi:10.1080/09553002.2017.123194427705054
  • Muxika A, Etxabide A, Uranga J, Guerrero P, de la Caba K. Chitosan as a bioactive polymer: processing, properties and applications. Int J Biol Macromol. 2017;105:1358–1368. doi:10.1016/j.ijbiomac.2017.07.08728735006
  • Younes I, Rinaudo M. Chitin and chitosan preparation from marine sources. Structure, properties and applications. Mar Drugs. 2015;13(3):1133–1174. doi:10.3390/md1303113325738328
  • Fathy MM, Mohamed FS, Elbialy N, Elshemey WM. Multifunctional chitosan-capped gold nanoparticles for enhanced cancer chemo-radiotherapy: an invitro study. Phys Med. 2018;48:76–83. doi:10.1016/j.ejmp.2018.04.00229728233
  • Cheung RC, Ng TB, Wong JH, Chan WY. Chitosan: an update on potential biomedical and pharmaceutical applications. Mar Drugs. 2015;13(8):5156–5186. doi:10.3390/md1308515626287217
  • Eblan MJ, Wang AZ. Improving chemoradiotherapy with nanoparticle therapeutics. Transl Cancer Res. 2013;2(4):320–329.25429359
  • Pearcey R, Brundage M, Drouin P, et al. Phase III trial comparing radical radiotherapy with and without cisplatin chemotherapy in patients with advanced squamous cell cancer of the cervix. J Clin Oncol. 2002;20(4):966–972. doi:10.1200/JCO.2002.20.4.96611844818
  • Chemoradiotherapy for Cervical Cancer Meta-Analysis Collaboration. Reducing uncertainties about the effects of chemoradiotherapy for cervical cancer: a systematic review and meta-analysis of individual patient data from 18 randomized trials. J Clin Oncol. 2008;26(35):5802–5812. doi:10.1200/JCO.2008.16.436819001332
  • Dong L, Li M, Zhang S, et al. Cytotoxicity of BSA-stabilized gold nanoclusters: in vitro and in vivo study. Small. 2015;11(21):2571–2581. doi:10.1002/smll.20140348125630756
  • Al-Jawad SMH, Taha AA, Al-Halbosiy MMF, Al-Barram LFA. Synthesis and characterization of small-sized gold nanoparticles coated by bovine serum albumin (BSA) for cancer photothermal therapy. Photodiagnosis Photodyn Ther. 2018;21:201–210. doi:10.1016/j.pdpdt.2017.12.00429223737
  • Murawala P, Tirmale A, Shiras A, Prasad BL. In situ synthesized BSA capped gold nanoparticles: effective carrier of anticancer drug methotrexate to MCF-7 breast cancer cells. Mater Sci Eng C Mater Biol Appl. 2014;34:158–167. doi:10.1016/j.msec.2013.09.00424268245
  • Kefayat A, Ghahremani F, Motaghi H, Mehrgardi MA. Investigation of different targeting decorations effect on the radiosensitizing efficacy of albumin-stabilized gold nanoparticles for breast cancer radiation therapy. Eur J Pharm Sci. 2019;130:225–233. doi:10.1016/j.ejps.2019.01.03730711685
  • Zhou B, Song J, Wang M, et al. BSA-bioinspired gold nanorods loaded with immunoadjuvant for the treatment of melanoma by combined photothermal therapy and immunotherapy. Nanoscale. 2018;10(46):21640–21647.30232481
  • Liu S, Piao J, Liu Y, et al. Radiosensitizing effects of different size bovine serum albumin-templated gold nanoparticles on H22 hepatoma-bearing mice. Nanomedicine (Lond). 2018;13(11):1371–1383. doi:10.2217/nnm-2018-005929749804
  • Koutcher L, Sherman E, Fury M, et al. Concurrent cisplatin and radiation versus cetuximab and radiation for locally advanced head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2011;81(4):915–922. doi:10.1016/j.ijrobp.2010.07.00820947269
  • Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharm. 2014;740:364–378. doi:10.1016/j.ejphar.2014.07.025
  • Setua S, Ouberai M, Piccirillo SG, Watts C, Welland M. Cisplatin-tethered gold nanospheres for multimodal chemo-radiotherapy of glioblastoma. Nanoscale. 2014;6(18):10865–10873. doi:10.1039/C4NR03693J25117686
  • Davidi ES, Dreifuss T, Motiei M, et al. Cisplatin-conjugated gold nanoparticles as a theranostic agent for head and neck cancer. Head Neck. 2018;40(1):70–78. doi:10.1002/hed.2493529130566
  • Cui L, Her S, Dunne M, et al. Significant radiation enhancement effects by gold nanoparticles in combination with cisplatin in triple negative breast cancer cells and tumor xenografts. Radiat Res. 2017;187(2):147–160. doi:10.1667/RR14578.128085639
  • Park J, Park J, Ju EJ, et al. Multifunctional hollow gold nanoparticles designed for triple combination therapy and CT imaging. J Control Release. 2015;207:77–85. doi:10.1016/j.jconrel.2015.04.00725863273
  • Rosenblum D, Joshi N, Tao W, Karp JM, Peer D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun. 2018;9(1):1410. doi:10.1038/s41467-018-03705-y29650952
  • Sinha R, Kim GJ, Nie S, Shin DM. Nanotechnology in cancer therapeutics: bioconjugated nanoparticles for drug delivery. Mol Cancer Ther. 2006;5(8):1909–1917. doi:10.1158/1535-7163.MCT-06-014116928810
  • Khoshgard K, Hashemi B, Arbabi A, Rasaee MJ, Soleimani M. Radiosensitization effect of folate-conjugated gold nanoparticles on HeLa cancer cells under orthovoltage superficial radiotherapy techniques. Phys Med Biol. 2014;59(9):2249–2263. doi:10.1088/0031-9155/59/9/224924733041
  • Samadian H, Hosseini-Nami S, Kamrava SK, Ghaznavi H, Shakeri-Zadeh A. Folate-conjugated gold nanoparticle as a new nanoplatform for targeted cancer therapy. J Cancer Res Clin Oncol. 2016;142(11):2217–2229. doi:10.1007/s00432-016-2179-327209529
  • Kefayat A, Ghahremani F, Motaghi H, Amouheidari A. Ultra-small but ultra-effective: folic acid-targeted gold nanoclusters for enhancement of intracranial glioma tumors’ radiation therapy efficacy. Nanomedicine. 2019;16:173–184. doi:10.1016/j.nano.2018.12.00730594659
  • Cruz E, Kayser V. Synthesis and enhanced cellular uptake in vitro of anti-HER2 multifunctional gold nanoparticles. Cancers. 2019;11(6):870. doi:10.3390/cancers11060870
  • Liu M, Yang YJ, Zheng H, et al. Membrane-bound complement regulatory proteins are prognostic factors of operable breast cancer treated with adjuvant trastuzumab: a retrospective study. Oncol Rep. 2014;32(6):2619–2627. doi:10.3892/or.2014.349625241923
  • Cai Z, Yook S, Lu Y, et al. Local radiation treatment of HER2-positive breast cancer using trastuzumab-modified gold nanoparticles labeled with Lu. Pharm Res. 2017;34(3):579–590. doi:10.1007/s11095-016-2082-227987070
  • Yook S, Cai Z, Lu Y, Winnik MA, Pignol JP, Reilly RM. Radiation nanomedicine for EGFR-positive breast cancer: panitumumab-modified gold nanoparticles complexed to the β-particle-emitter, (177)Lu. Mol Pharm. 2015;12(11):3963–3972. doi:10.1021/acs.molpharmaceut.5b0042526402157
  • Yook S, Cai Z, Lu Y, Winnik MA, Pignol JP, Reilly RM. Intratumorally injected 177Lu-labeled gold nanoparticles: gold nanoseed brachytherapy with application for neoadjuvant treatment of locally advanced breast cancer. J Nucl Med. 2016;57(6):936–942. doi:10.2967/jnumed.115.16890626848176
  • Hatoyama K, Kitamura N, Takano-Kasuya M, et al. Quantitative analyses of amount and localization of radiosensitizer gold nanoparticles interacting with cancer cells to optimize radiation therapy. Biochem Biophys Res Commun. 2019;508(4):1093–1100. doi:10.1016/j.bbrc.2018.12.01630551875
  • Popovtzer A, Mizrachi A, Motiei M, et al. Actively targeted gold nanoparticles as novel radiosensitizer agents: an in vivo head and neck cancer model. Nanoscale. 2016;8(5):2678–2685. doi:10.1039/C5NR07496G26757746
  • Jóźwiak P, Lipińska A. The role of glucose transporter 1 (GLUT1) in the diagnosis and therapy of tumors. Postepy Hig Med Dosw. 2012;66:165–174. (Article in Polish).
  • Song L, Falzone N, Vallis KA. EGF-coated gold nanoparticles provide an efficient nano-scale delivery system for the molecular radiotherapy of EGFR-positive cancer. Int J Radiat Biol. 2016;92(11):716–723. doi:10.3109/09553002.2016.114536026999580
  • Barron CC, Bilan PJ, Tsakiridis T, Tsiani E. Facilitative glucose transporters: implications for cancer detection, prognosis and treatment. Metabolism. 2016;65(2):124–139. doi:10.1016/j.metabol.2015.10.00726773935
  • Calvo MB, Figueroa A, Pulido EG, Campelo RG, Aparicio LA. Potential role of sugar transporters in cancer and their relationship with anticancer therapy. Int J Endocrinol. 2010;2010:205357. doi:10.1155/2010/20535720706540
  • Kong T, Zeng J, Wang X, et al. Enhancement of radiation cytotoxicity in breast-cancer cells by localized attachment of gold nanoparticles. Small. 2008;4(9):1537–1543. doi:10.1002/smll.20070079418712753
  • Roa W, Zhang X, Guo L, et al. Gold nanoparticle sensitize radiotherapy of prostate cancer cells by regulation of the cell cycle. Nanotechnology. 2009;20(37):375101. doi:10.1088/0957-4484/20/37/37510119706948
  • Wang C, Jiang Y, Li X, Hu L. Thioglucose-bound gold nanoparticles increase the radiosensitivity of a triple-negative breast cancer cell line (MDA-MB-231). Breast Cancer. 2015;22(4):413–420. doi:10.1007/s12282-013-0496-924114595
  • Soleymanifard S, Rostami A, Aledavood SA, Matin MM, Sazgarnia A. Increased radiotoxicity in two cancerous cell lines irradiated by low and high energy photons in the presence of thio-glucose bound gold nanoparticles. Int J Radiat Biol. 2017;93(4):407–415. doi:10.1080/09553002.2017.126828227921518
  • Hu C, Niestroj M, Yuan D, Chang S, Chen J. Treating cancer stem cells and cancer metastasis using glucose-coated gold nanoparticles. Int J Nanomed. 2015;10:2065–2077.
  • Park EI, Manzella SM, Baenziger JU. Rapid clearance of sialylated glycoproteins by the asialoglycoprotein receptor. J Biol Chem. 2003;278(7):4597–4602. doi:10.1074/jbc.M21061220012464602
  • Zhu CD, Zheng Q, Wang LX, et al. Synthesis of novel galactose functionalized gold nanoparticles and its radiosensitizing mechanism. J Nanobiotechnol. 2015;13:67. doi:10.1186/s12951-015-0129-x
  • Kunjachan S, Pola R, Gremse F, et al. Passive versus active tumor targeting using RGD- and NGR-modified polymeric nanomedicines. Nano Lett. 2014;14(2):972–981. doi:10.1021/nl404391r24422585
  • Ruoslahti E, Pierschbacher MD. New perspectives in cell adhesion: RGD and integrins. Science. 1987;238(4826):491–497. doi:10.1126/science.28216192821619
  • Wu PH, Onodera Y, Ichikawa Y, et al. Targeting integrins with RGD-conjugated gold nanoparticles in radiotherapy decreases the invasive activity of breast cancer cells. Int J Nanomed. 2017;12:5069–5085. doi:10.2147/IJN.S137833
  • Özcelik S, Pratx G. Nuclear-targeted gold nanoparticles enhance cancer cell radiosensitization. Nanotechnology. 2020;31(41):415102. doi:10.1088/1361-6528/aba02b32585647
  • Kunjachan S, Detappe A, Kumar R, et al. Nanoparticle mediated tumor vascular disruption: a novel strategy in radiation therapy. Nano Lett. 2015;15(11):7488–7496. doi:10.1021/acs.nanolett.5b0307326418302
  • Banerjee D, Harfouche R, Sengupta S. Nanotechnology-mediated targeting of tumor angiogenesis. Vasc Cell. 2011;3(1):3. doi:10.1186/2045-824X-3-321349160
  • Yang CJ, Chithrani DB. Nuclear targeting of gold nanoparticles for improved therapeutics. Curr Top Med Chem. 2016;16(3):271–280. doi:10.2174/156802661566615070111501226126911
  • Farkhani SM, Valizadeh A, Karami H, Mohammadi S, Sohrabi N, Badrzadeh F. Cell penetrating peptides: efficient vectors for delivery of nanoparticles, nanocarriers, therapeutic and diagnostic molecules. Peptides. 2014;57:78–94. doi:10.1016/j.peptides.2014.04.01524795041
  • Kalmouni M, Al-Hosani S, Magzoub M. Cancer targeting peptides. Cell Mol Life Sci. 2019;76(11):2171–2183.30877335
  • Ramsey JD, Flynn NH. Cell-penetrating peptides transport therapeutics into cells. Pharmacol Ther. 2015;154:78–86. doi:10.1016/j.pharmthera.2015.07.00326210404
  • Wang F, Wang Y, Zhang X, Zhang W, Guo S, Jin F. Recent progress of cell-penetrating peptides as new carriers for intracellular cargo delivery. J Control Release. 2014;174:126–136. doi:10.1016/j.jconrel.2013.11.02024291335
  • Zhang X, Wang H, Coulter JA, Yang R. Octaarginine-modified gold nanoparticles enhance the radiosensitivity of human colorectal cancer cell line LS180 to megavoltage radiation. Int J Nanomed. 2018;13:3541–3552. doi:10.2147/IJN.S161157
  • Yang C, Neshatian M, van Prooijen M. Cancer nanotechnology: enhanced therapeutic response using peptide-modified gold nanoparticles. J Nanosci Nanotechnol. 2014;14(7):4813–4819. doi:10.1166/jnn.2014.928024757948
  • Yang C, Uertz J, Yohan D, Chithrani BD. Peptide modified gold nanoparticles for improved cellular uptake, nuclear transport, and intracellular retention. Nanoscale. 2014;6(20):12026–12033. doi:10.1039/C4NR02535K25182693
  • Mackey MA, El-Sayed MA. Chemosensitization of cancer cells via gold nanoparticle-induced cell cycle regulation. Photochem Photobiol. 2014;90(2):306–312. doi:10.1111/php.1222624329577
  • Bates PJ, Laber DA, Miller DM, Thomas SD, Trent JO. Discovery and development of the G-rich oligonucleotide AS1411 as a novel treatment for cancer. Exp Mol Pathol. 2009;86(3):151–164. doi:10.1016/j.yexmp.2009.01.00419454272
  • Zhu G, Chen X. Aptamer-based targeted therapy. Adv Drug Deliv Rev. 2018;134:65–78. doi:10.1016/j.addr.2018.08.00530125604
  • Morita Y, Leslie M, Kameyama H, Volk DE, Tanaka T. Aptamer therapeutics in cancer: current and future. Cancers (Basel). 2018;10(3):80. doi:10.3390/cancers10030080
  • Soundararajan S, Chen W, Spicer EK, Courtenay-Luck N, Fernandes DJ. The nucleolin targeting aptamer AS1411 destabilizes Bcl-2 messenger RNA in human breast cancer cells. Cancer Res. 2008;68(7):2358–2365. doi:10.1158/0008-5472.CAN-07-572318381443
  • Li X, Wang H, Lu X, Di, B. Silencing STAT3 with short hairpin RNA enhances radiosensitivity of human laryngeal squamous cell carcinoma xenografts in vivo. Exp Ther Med. 2010;1(6):947–953.22993624
  • Geiger JL, Grandis JR, Bauman JE. The STAT3 pathway as a therapeutic target in head and neck cancer: barriers and innovations. Oral Oncol. 2016;56:84–92. doi:10.1016/j.oraloncology.2015.11.02226733183
  • Oweida AJ, Darragh L, Phan A, et al. STAT3 modulation of regulatory T cells in response to radiation therapy in head and neck cancer. J Natl Cancer Inst. 2019;111(12):1339–1349. doi:10.1093/jnci/djz03630863843
  • Zhang S, Gupta S, Fitzgerald TJ, Bogdanov AA. Dual radiosensitization and anti-STAT3 anti-proliferative strategy based on delivery of gold nanoparticle - oligonucleotide nanoconstructs to head and neck cancer cells. Nanotheranostics. 2018;2(1):1–11. doi:10.7150/ntno.2233529291159
  • Luo CH, Huang CT, Su CH, Yeh CS. Bacteria-mediated hypoxia-specific delivery of nanoparticles for tumors imaging and therapy. Nano Lett. 2016;16(6):3493–3499. doi:10.1021/acs.nanolett.6b0026227148804
  • Roma-Rodrigues C, Pombo I, Raposo L, Pedrosa P, Fernandes AR, Baptista PV. Nanotheranostics targeting the tumor microenvironment. Front Bioeng Biotechnol. 2019;7:197.31475143
  • Zhao Z, Xu H, Li S, et al. Hypoxic radiosensitizer-lipid coated gold nanoparticles enhance the effects of radiation therapy on tumor growth. J Biomed Nanotechnol. 2019;15(9):1982–1993. doi:10.1166/jbn.2019.283031387684
  • Wang Y, Shang W, Niu M, Tian J, Xu K. Hypoxia-active nanoparticles used in tumor theranostic. Int J Nanomed. 2019;14:3705–3722. doi:10.2147/IJN.S196959
  • Chen Q, Feng L, Liu J, et al. Intelligent albumin-MnO2 nanoparticles as pH-/H2O2 -responsive dissociable nanocarriers to modulate tumor hypoxia for effective combination therapy. Adv Mater. 2016;28(33):7129–7136. doi:10.1002/adma.20160190227283434
  • Ding Y, Sun Z, Tong Z, et al. Tumor microenvironment-responsive multifunctional peptide coated ultrasmall gold nanoparticles and their application in cancer radiotherapy. Theranostics. 2020;10(12):5195–5208. doi:10.7150/thno.4501732373207
  • Zhang Y, Yang L, Yang C, Liu J. Recent advances of smart acid-responsive gold nanoparticles in tumor therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2020;12(4):e1619. doi:10.1002/wnan.161932043312
  • Yao L, Daniels J, Moshnikova A, et al. pHLIP peptide targets nanogold particles to tumors. Proc Natl Acad Sci U S A. 2013;110(2):465–470. doi:10.1073/pnas.121966511023267062
  • Daniels JL, Crawford TM, Andreev OA, Reshetnyak YK. Synthesis and characterization of pHLIP coated gold nanoparticles. Biochem Biophys Rep. 2017;10:62–69.28955736
  • Antosh MP, Wijesinghe DD, Shrestha S, et al. Enhancement of radiation effect on cancer cells by gold-pHLIP. Proc Natl Acad Sci U S A. 2015;112(17):5372–5376. doi:10.1073/pnas.150162811225870296
  • Horsman MR, Overgaard J. The impact of hypoxia and its modification of the outcome of radiotherapy. J Radiat Res. 2016;Suppl 1:i90–i98. doi:10.1093/jrr/rrw007
  • Martin JD, Fukumura D, Duda DG, Boucher Y, Jain RK. Reengineering the tumor microenvironment to alleviate hypoxia and overcome cancer heterogeneity. Cold Spring Harb Perspect Med. 2016;6(12):a027094. doi:10.1101/cshperspect.a02709427663981
  • Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15(7):409–425. doi:10.1038/nrc395826105538
  • Xie J, Gong L, Zhu S, Yong Y, Gu Z, Zhao Y. Emerging strategies of nanomaterial-mediated tumor radiosensitization. Adv Mater. 2019;31(3):e1802244. doi:10.1002/adma.20180224430156333
  • Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci. 2012;33(4):207–214. doi:10.1016/j.tips.2012.01.00522398146
  • Schito L, Semenza GL. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer. 2016;2(12):758–770. doi:10.1016/j.trecan.2016.10.01628741521
  • Patiar S, Harris AL. Role of hypoxia-inducible factor-1alpha as a cancer therapy target. Endocr Relat Cancer. 2006;Suppl 1:S61–S75. doi:10.1677/erc.1.01290
  • Kim MS, Lee EJ, Kim JW, et al. Gold nanoparticles enhance anti-tumor effect of radiotherapy to hypoxic tumor. Radiat Oncol J. 2016;34(3):230–238. doi:10.3857/roj.2016.0178827730800
  • Gong F, Yang N, Wang X, et al. Tumor microenvironment-responsive intelligent nanoplatforms for cancer theranostics. Nano Today. 2020;32:100851. doi:10.1016/j.nantod.2020.100851
  • Li J, Shang W, Li Y, Fu S, Tian J, Lu L. Advanced nanomaterials targeting hypoxia to enhance radiotherapy. Int J Nanomed. 2018;13:5925. doi:10.2147/IJN.S173914
  • Kefayat A, Ghahremani F, Motaghi H, Rostami S, Mehrgardi MA. Alive attenuated Salmonella as a cargo shuttle for smart carrying of gold nanoparticles to tumour hypoxic regions. J Drug Target. 2019;27(3):315–324. doi:10.1080/1061186X.2018.152341730207745
  • Cho WS, Cho M, Jeong J, et al. Size-dependent tissue kinetics of PEG-coated gold nanoparticles. Toxicol Appl Pharmacol. 2010;245(1):116–123. doi:10.1016/j.taap.2010.02.01320193702
  • Chou LY, Chan WC. Fluorescence‐tagged gold nanoparticles for rapidly characterizing the size‐dependent biodistribution in tumor models. Adv Healthc Mater. 2012;1(6):714–721.23184822
  • Li B, Lane LA. Probing the biological obstacles of nanomedicine with gold nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11(3):e1542.30084539
  • Bai X, Liu F, Liu Y, et al. Toward a systematic exploration of nano-bio interactions. Toxicol Appl Pharmacol. 2017;323:66–73. doi:10.1016/j.taap.2017.03.01128344110
  • Abdulle A, Chow JCL. Contrast enhancement for portal imaging in nanoparticle-enhanced radiotherapy: a monte carlo phantom evaluation using flattening-filter-free photon beams. Nanomaterials (Basel). 2019;9(7):920. doi:10.3390/nano9070920
  • Albayedh F, Chow JCL. Monte carlo simulation on the imaging contrast enhancement in nanoparticle-enhanced radiotherapy. J Med Phys. 2018;43(3):195–199.30305778
  • Dimitriou NM, Tsekenis G, Balanikas EC, et al. Gold nanoparticles, radiations and the immune system: current insights into the physical mechanisms and the biological interactions of this new alliance towards cancer therapy. Pharmacol Ther. 2017;178:1–17. doi:10.1016/j.pharmthera.2017.03.00628322970
  • Batooei S, Khajeali A, Khodadadi R, Islamian JP. Metal-based nanoparticles as radio-sensitizer in gastric cancer therapy. J Drug Deliv Sci Technol. 2020;56:101576. doi:10.1016/j.jddst.2020.101576
  • Butterworth KT, McMahon SJ, Currell FJ, Prise KM. Physical basis and biological mechanisms of gold nanoparticle radiosensitization. Nanoscale. 2012;4(16):4830–4838. doi:10.1039/c2nr31227a22767423
  • Rosa S, Connolly C, Schettino G, Butterworth KT, Prise KM. Biological mechanisms of gold nanoparticle radiosensitization. Cancer Nanotechnol. 2017;8(1):2. doi:10.1186/s12645-017-0026-028217176
  • Retif P, Pinel S, Toussaint M, et al. Nanoparticles for radiation therapy enhancement: the key parameters. Theranostics. 2015;5(9):1030–1044. doi:10.7150/thno.1164226155318
  • Yao X, Huang C, Chen X, Yi Z, Sanche L. Chemical radiosensitivity of DNA induced by gold nanoparticles. J Biomed Nanotechnol. 2015;11(3):478–485. doi:10.1166/jbn.2015.192226307830
  • Liu K, Han L, Zhuang J, Yang DP. Protein-directed gold nanoparticles with excellent catalytic activity for 4-nitrophenol reduction. Mater Sci Eng C Mater Biol Appl. 2017;78:429–434. doi:10.1016/j.msec.2017.04.05228576005
  • Paunesku T, Gutiontov S, Brown K, Woloschak GE. Radiosensitization and nanoparticles. Cancer Treat Res. 2015;166:151–171.25895868
  • Cui L, Her S, Borst GR, Bristow RG, Jaffray DA, Allen C. Radiosensitization by gold nanoparticles: will they ever make it to the clinic? Radiother Oncol. 2017;124(3):344–356. doi:10.1016/j.radonc.2017.07.00728784439
  • Pateras IS, Havaki S, Nikitopoulou X, et al. The DNA damage response and immune signaling alliance: is it good or bad? Nature decides when and where. Pharmacol Ther. 2015;154:36–56. doi:10.1016/j.pharmthera.2015.06.01126145166
  • Martelli S, Chow JCL. Dose enhancement for the flattening-filter-free and flattening-filter photon beams in nanoparticle-enhanced radiotherapy: a Monte Carlo phantom Study. Nanomaterials (Basel). 2020;10(4):637. doi:10.3390/nano10040637
  • Dewmini Mututantri-Bastiyange JC, Chow L. Imaging dose of cone-beam computed tomography in nanoparticle-enhanced image-guided radiotherapy: a Monte Carlo phantom study. AIMS Bioeng. 2020;7(1):1–11. doi:10.3934/bioeng.2020001
  • Cho SH. Estimation of tumour dose enhancement due to gold nanoparticles during typical radiation treatments: a preliminary Monte Carlo study. Phys Med Biol. 2005;50(15):N163–N173. doi:10.1088/0031-9155/50/15/N0116030374
  • Chow JC, Leung MK, Jaffray DA. Monte Carlo simulation on a gold nanoparticle irradiated by electron beams. Phys Med Biol. 2012;57(11):3323–3331. doi:10.1088/0031-9155/57/11/332322572475
  • Jain S, Coulter JA, Hounsell AR, et al. Cell-specific radiosensitization by gold nanoparticles at megavoltage radiation energies. Int J Radiat Oncol Biol Phys. 2011;79(2):531–539. doi:10.1016/j.ijrobp.2010.08.04421095075
  • Lin Y, McMahon SJ, Scarpelli M, Paganetti H, Schuemann J. Comparing gold nano-particle enhanced radiotherapy with protons, megavoltage photons and kilovoltage photons: a Monte Carlo simulation. Phys Med Biol. 2014;59(24):7675–7689. doi:10.1088/0031-9155/59/24/767525415297
  • Jeynes JC, Merchant MJ, Spindler A, Wera AC, Kirkby KJ. Investigation of gold nanoparticle radiosensitization mechanisms using a free radical scavenger and protons of different energies. Phys Med Biol. 2014;59(21):6431–6443. doi:10.1088/0031-9155/59/21/643125296027
  • Cui L, Tse K, Zahedi P, et al. Hypoxia and cellular localization influence the radiosensitizing effect of gold nanoparticles (AuNPs) in breast cancer cells. Radiat Res. 2014;182(5):475–488. doi:10.1667/RR13642.125361396
  • Mkandawire MM, Lakatos M, Springer A, et al. Induction of apoptosis in human cancer cells by targeting mitochondria with gold nanoparticles. Nanoscale. 2015;7(24):10634–10640. doi:10.1039/C5NR01483B26022234
  • Yang Y, Ren L, Wang H. Strategies in the design of gold nanoparticles for intracellular targeting: opportunities and challenges. Ther Deliv. 2017;8(10):879–897. doi:10.4155/tde-2017-004928944735
  • Pathak RK, Kolishetti N, Dhar S. Targeted nanoparticles in mitochondrial medicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2015;7(3):315–329. doi:10.1002/wnan.130525348382
  • Mateo D, Morales P, Ávalos A, Haza AI. Oxidative stress contributes to gold nanoparticle-induced cytotoxicity in human tumor cells. Toxicol Mech Methods. 2014;24(3):161–172. doi:10.3109/15376516.2013.86978324274460
  • Decrock E, Hoorelbeke D, Ramadan R, et al. Calcium, oxidative stress and connexin channels, a harmonious orchestra directing the response to radiotherapy treatment? Biochim Biophys Acta Mol Cell Res. 2017;1864(6):1099–1120. doi:10.1016/j.bbamcr.2017.02.00728193563
  • Liu R, Wang Y, Yuan Q, An D, Li J, Gao X. The Au clusters induce tumor cell apoptosis via specifically targeting thioredoxin reductase 1 (TrxR1) and suppressing its activity. Chem Commun (Camb). 2014;50(73):10687–10690. doi:10.1039/C4CC03320E25078326
  • Pawlik TM, Keyomarsi K. Role of cell cycle in mediating sensitivity to radiotherapy. Int J Radiat Oncol Biol Phys. 2004;59(4):928–942. doi:10.1016/j.ijrobp.2004.03.00515234026
  • Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 2004;432(7015):316–323. doi:10.1038/nature0309715549093
  • Mackey MA, Saira F, Mahmoud MA, El-Sayed MA. Inducing cancer cell death by targeting its nucleus: solid gold nanospheres versus hollow gold nanocages. Bioconjug Chem. 2013;24(6):897–906. doi:10.1021/bc300592d23777334
  • Pan Y, Leifert A, Ruau D, et al. Gold nanoparticles of diameter 1.4 nm trigger necrosis by oxidative stress and mitochondrial damage. Small. 2009;5(18):2067–2076. doi:10.1002/smll.20090046619642089
  • Butterworth KT, Coulter JA, Jain S, et al. Evaluation of cytotoxicity and radiation enhancement using 1.9 nm gold particles: potential application for cancer therapy. Nanotechnology. 2010;21(29):295101. doi:10.1088/0957-4484/21/29/29510120601762
  • Djuzenova CS, Elsner I, Katzer A, et al. Radiosensitivity in breast cancer assessed by the histone γ-H2AX and 53BP1 foci. Radiat Oncol. 2013;8:98. doi:10.1186/1748-717X-8-9823617930
  • Vignard J, Mirey G, Salles B. Ionizing-radiation induced DNA double-strand breaks: a direct and indirect lighting up. Radiother Oncol. 2013;108(3):362–369. doi:10.1016/j.radonc.2013.06.01323849169
  • Galluzzi L, Pietrocola F, Bravo-San Pedro JM, et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015;34(7):856–880. doi:10.15252/embj.20149078425712477
  • Yang A, Rajeshkumar NV, Wang X, et al. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 2014;4(8):905–913. doi:10.1158/2159-8290.CD-14-036224875860
  • Xu DW, Zhang GQ, Wang ZW, Xu XY, Liu TX. Autophagy in tumorigenesis and cancer treatment. Asian Pac J Cancer Prev. 2015;16(6):2167–2175. doi:10.7314/APJCP.2015.16.6.216725824733
  • Towers CG, Thorburn A. Therapeutic targeting of autophagy. EBioMedicine. 2016;14:15–23. doi:10.1016/j.ebiom.2016.10.03428029600
  • Ma X, Wu Y, Jin S, et al. Gold nanoparticles induce autophagosome accumulation through size-dependent nanoparticle uptake and lysosome impairment. ACS Nano. 2011;5(11):8629–8639. doi:10.1021/nn202155y21974862
  • Verfaillie T, Garg AD, Agostinis P. Targeting ER stress induced apoptosis and inflammation in cancer. Cancer Lett. 2013;332(2):249–264. doi:10.1016/j.canlet.2010.07.01620732741
  • Bhat TA, Chaudhary AK, Kumar S, et al. Endoplasmic reticulum-mediated unfolded protein response and mitochondrial apoptosis in cancer. Biochim Biophys Acta Rev Cancer. 2017;1867(1):58–66. doi:10.1016/j.bbcan.2016.12.00227988298
  • Cubillos-Ruiz JR, Bettigole SE, Glimcher LH. Tumorigenic and Immunosuppressive effects of endoplasmic reticulum stress in cancer. Cell. 2017;168(4):692–706. doi:10.1016/j.cell.2016.12.00428187289
  • Mohamed E, Cao Y, Rodriguez PC. Endoplasmic reticulum stress regulates tumor growth and anti-tumor immunity: a promising opportunity for cancer immunotherapy. Cancer Immunol Immunother. 2017;66(8):1069–1078.28577085
  • Tsai YY, Huang YH, Chao YL, et al. Identification of the nanogold particle-induced endoplasmic reticulum stress by omic techniques and systems biology analysis. ACS Nano. 2011;5(12):9354–9369. doi:10.1021/nn202777522107733
  • Simard JC, Durocher I, Girard D. Silver nanoparticles induce irremediable endoplasmic reticulum stress leading to unfolded protein response dependent apoptosis in breast cancer cells. Apoptosis. 2016;21(11):1279–1290. doi:10.1007/s10495-016-1285-727586505
  • Chen R, Huo L, Shi X, et al. Endoplasmic reticulum stress induced by zinc oxide nanoparticles is an earlier biomarker for nanotoxicological evaluation. ACS Nano. 2014;8(3):2562–2574. doi:10.1021/nn406184r24490819
  • Yang X, Shao H, Liu W, et al. Endoplasmic reticulum stress and oxidative stress are involved in ZnO nanoparticle-induced hepatotoxicity. Toxicol Lett. 2015;234(1):40–49. doi:10.1016/j.toxlet.2015.02.00425680694
  • Le Sech C, Kobayashi K, Usami N, Furusawa Y, Porcel E, Lacombe S. Comment on therapeutic application of metallic nanoparticles combined with particle-induced x-ray emission effect. Nanotechnology. 2012;23(7):078001. doi:10.1088/0957-4484/23/7/07800122261552
  • Koonce NA, Quick CM, Hardee ME, et al. Combination of gold nanoparticle-conjugated tumor necrosis factor-α and radiation therapy results in a synergistic antitumor response in murine carcinoma models. Int J Radiat Oncol Biol Phys. 2015;93(3):588–596. doi:10.1016/j.ijrobp.2015.07.227526461001
  • Khoobchandani M, Katti KK, Karikachery AR, et al. New approaches in breast cancer therapy through green nanotechnology and nano-ayurvedic medicine - pre-clinical and pilot human clinical investigations. Int J Nanomed. 2020;15:181–197. doi:10.2147/IJN.S219042
  • Lawrence YR, Vikram B, Dignam JJ, et al. NCI-RTOG translational program strategic guidelines for the early-stage development of radiosensitizers. J Natl Cancer Inst. 2013;105(1):11–24. doi:10.1093/jnci/djs47223231975