AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2.9 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Metal, Metalloid, and Oxide Nanoparticles for Therapeutic and Diagnostic Oncology

Mohammad Hossein Yazdi1,3Zargham Sepehrizadeh1Mehdi Mahdavi2,3Ahmad Reza Shahverdi1,3( )Mohammad Ali Faramarzi1( )
Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
Recombinant Vaccine Research Center, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
Show Author Information

Abstract

Nanoparticles have comprehensively affected various sights of human life. Through the wide range of claims about nanosized particles and their functions, biomedical applications are of much interest among health care researchers due to the nanoparticles' potential for use in the process of disease diagnosis, control and treatment. In this regard, inorganic nanoparticles, which have high potential in diagnostic and therapeutic systems, have recently received much attention in oncology. Although inorganic nanoparticles initially seemed an appropriate tool for cancer imaging and diagnosis, their ability to attack cancerous cells as anticancer drugs or carriers in other drugs has also demonstrated promising results. The present review primarily provides a brief survey of various studies in which metal nanoparticles such as gold, silver, iron oxide, and metalloid nanoparticles viz. tellurium and bismuth were exploited for therapeutic or diagnostic purposes in oncology. Then the application of selenium nanoparticles as a therapeutic agent against cancer in in vitro and in vivo studies is reviewed in detail. Although inorganic nanoparticles seem to be useful tools for cancer imaging and diagnosis, their potential for attacking cancerous cells as anticancer substances or even carriers for anticancer medications should not be underestimated.

References

[1]

T.K. Sau, A.L. Rogach, Nonspherical noble metal NPs: colloid-chemical synthesis and morphology control. Advanced Materials, 2010, 22:1781-1804.

[2]

H. Schmidt, NPs by chemical synthesis, processing to materials and innovative applications. Applied Organometallic Chemistry, 2001, 15:331-343.

[3]

A.B.D. Pandey. Synthesis of zinc-based nanomaterials: a biological perspective. IET Nanobiotechnology, 2012, 6:144-148.

[4]

M. Shakibaie, H. Forootanfar, K.M. Moghaddam, et al., Green synthesis of gold NPs by the marine microalga Tetraselmis suecica. Biotechnology and Applied Biochemistry, 2010, 57:71-75.

[5]

B. Kang, M.A. Mackey, M.A. El-Sayed, Nuclear targeting of gold NPs in cancer cells induces DNA damage, causing cytokinesis arrest and apoptosis. Journal of the American Chemical Society, 2010, 132:1517-1519.

[6]

M.M. Yallapu, S.P. Foy, T.K. Jain, et al., PEG-functionalized magnetic NPs for drug delivery and magnetic resonance imaging applications. Pharmacological Research, 2010, 27:2283-2295.

[7]

P.R. Gil, D. Huhn, L.L. Del Mercato, et al., Nanopharmacy: Inorganic nanoscale devices as vectors and active compounds. Pharmacological Research , 2010, 62:115-125.

[8]

S. Bhattacharyya, R.A. Kudgus, R. Bhattacharya, et al., Inorganic NPs in cancer therapy. Pharmacological Research, 2011, 28:237-59.

[9]

N.L. Rosi, C.A. Mirkin, Nanostructures in biodiagnostics. Chemical Reviews, 2005, 105:1547-1562.

[10]

H.C. Huang, S. Barua, G. Sharma, et al., Inorganic NPs for cancer imaging and therapy. Journal of Control Release, 2011, 155: 344-357.

[11]

K.H. Bae, H.J. Chung, and T.G. Park, Nanomaterials for Cancer Therapy and Imaging. Molecules and Cells, 2011, 31:295-302.

[12]

M. Valden, X. Lal, D.W. Goodman, Onset of catalytic activity of gold clusters on titania with the appearance of nonmetallic properties. Science, 1998, 281:1647-1650.

[13]

M.A. El-Sayed, Some interesting properties of metals confined in time and nanometer space of different shapes. Accounts of Chemical Research, 2001, 34:257-264.

[14]
D.L. Feldheim, C.A. Foss Jr., Metal NPs: synthesis characterization, and applications. Marcel Dekker, 2002.
[15]

C.J. Murphy, Nanocubes and Nanoboxes. Science, 2002, 298: 2139-2141.

[16]

S.T. Selvan, T.T. Tan, D.K. Yi, et al., Functional and multifunctional nanoparticles for bioimaging and biosensing. Langmuir, 2010, 26: 11631-11641.

[17]

Y. Liu, M. Xu, Q. Chen, et al., Gold nanorods/mesoporous silica-based nanocomposite as theranostic agents for targeting near-infrared imaging and photothermal therapy induced with laser. International Journal of Nanomedicine, 2015, 10: 4747-4761.

[18]

X. Huang, P.K. Jain, I.H. El−Sayed, et al., Gold NPs: interesting optical properties and recent applications in cancer diagnostics and therapy. Nanomedicine, 2007, 2: 681-693.

[19]

J.F. Hainfeld, D.N. Slatkin, and H.M. Smilowitz, The use of gold NPs to enhance radiotherapy in mice. Physics in Medicine and Biology, 2004, 49: 309-315.

[20]

J.F. Hainfeld, F.A. Dilmanian, D.N. Slatkin, et al., Radiotherapy enhancement with gold NPs. Journal of Pharmacy and Pharmacology, 2008, 60: 977-985.

[21]

L.R. Hirsch, R.J. Stafford, J.A. Bankson, et al., Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance. Proceedings of the National Academy of Sciences U.S.A., 2003, 100: 13549-13554.

[22]

X. Huang, I.H. El-Sayed, W. Qian, et al., Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. Journal of the American Chemical Society, 2006, 128: 2115-2120.

[23]

M.S. Yavuz, Y. Cheng, J. Chen, et al., Gold nanocages covered by smart polymers for controlled release with near-infrared light. Nature Materials, 2009, 8: 935-939.

[24]

G. Han, P. Ghosh, and V.M. Rotello, Multi-functional gold NPs for drug delivery. Advances in Experimental Medicine and Biology, 2007, 620: 48-56.

[25]

M. Thomas, A.M. Klibanov, Conjugation to gold NPs enhances polyethylenimine's transfer of plasmid DNA into mammalian cells. Proceedings of the National Academy of Sciences U.S.A, 2003, 100: 9138-9143.

[26]

Z.Yang , D. Gao , Z. Cao, et al., Drug and gene co-delivery systems for cancer treatment. Biomaterial Science, 2015, 3: 1035-1049.

[27]

A.S. Arbab, W. Liu, and J.A. Frank. Cellular magnetic resonance imaging: current status and future prospects. Expert Review of Medical Devices, 2006, 3: 427-439.

[28]

K. Kan-Dapaah, N. Rahbar, and W. Soboyejo, Novel magnetic heating probe for multimodal cancer treatment. Medical Physics, 2015, 42: 2203-2211.

[29]

T.K. Sau, A.L. Rogach, Nonspherical noble metal NPs: colloid-chemical synthesis and morphology control. Advanced Materials, 2010, 22: 1781-1804.

[30]

S. Patra, S. Mukherjee, A.K. Barui, et al., Green synthesis, characterization of gold and silver NPs and their potential application for cancer therapeutics. Materials Science and Engineering C: Materials for Biological Applications, 2015, 53: 298-309.

[31]

R.A. Sperling, W.J. Parak, Surface modification, functionalization and bioconjugation of colloidal inorganic NPs. Philosophical Transactions of the Royal Society A, 2010, 368: 1333-1383.

[32]

E. Katz, I. Willner, Integrated nanoparticle–biomolecule hybrid systems: synthesis, properties, and applications. Angewandte Chemie International Edition, 2004, 43: 6042-6108.

[33]

C.M. Niemeyer, NPs, proteins, and nucleic acids: biotechnology meets materials science. Angewandte Chemie International Edition, 2001, 40: 4128-4158.

[34]

K. Naka, H. Itoh, Y. Tampo, et al., Effect of gold NPs as a support for the oligomerization of L-cysteine in an aqueous solution. Langmuir, 2003, 19: 5546-5549.

[35]

P. Ghosh, G. Han, M. De, et al., Gold NPs in delivery applications. Advanced Drug Delivery Reviews, 2008, 60: 1307-1315.

[36]

N. Nishiyama, Nanomedicine: nanocarriers shape up for long life. Nature Nanotechnology, 2007, 2: 203-204.

[37]

H. Chen, L. Shao, T. Ming, et al., Understanding the photothermal conversion efficiency of gold nanocrystals. Small, 2010, 6: 2272-2280.

[38]

E.S. Day, J.G. Morton, and J.L. West, NPs for thermal cancer therapy. Journal of Biomechanical Engineering, 2009, 131: 740011.

[39]

K.K. Jain, Applications of nanobiotechnology in clinical diagnostics. Clinical Chemistry, 2007, 53: 2002-2009.

[40]

K.K. Jain, Nanomedicine: application of nanobiotechnology in medical practice. Medical Principles and Practice, 2008, 17: 89-101.

[41]

P.V. Baptista, G. Doria, P. Quaresma, et al., NPs in molecular diagnostics. Progress in Molecular Biology and Translational Science. 2011, 104: 427-488.

[42]

C. Jianrong, M. Yuqing, H. Nongyue, et al., Nanotechnology and biosensors. Biotechnology Advances, 2004, 22: 505-518.

[43]

B.S. Sekhon, Metalloid compounds as drugs. Research in Pharmaceutical Sciences, 2013, 8: 145-158.

[44]

N.C. Loyd, H.W. Morgan, B.K. Nicholson, et al., The composition of Ehrlich's salvarsan: resolution of a century-old debate. Angewandte Chemie International Edition in English, 2005, 44: 941-944.

[45]

S. Soflaei, A. Dalimi, F. Ghaffarifar, et al., In vitro antiparasitic and apoptotic effects of antimony sulfide NPs on Leishmania infantum. Journal of Parasitology Research, 2012, 756568.

[46]

R.L. Cunha, I.E. Gouvea, and L. Juliano, A glimpse on biological activities of tellurium compounds. Anais da Academia Brasileira de Ciências, 2009, 81: 393-407.

[47]

Z.P. Xu, Q.H. Zeng, G.Q. Lu, et al., Inorganic NPs as carriers for efficient cellular delivery. Chemical Engineering Science, 2006, 61: 1027-1040.

[48]

K. Byrappa, S. Ohara, and T. Adschiri, NPs synthesis using supercritical fluid technology - towards biomedical applications. Advanced Drug Delivery Reviews, 2008, 60: 299-307.

[49]

O. Rabin, J.M. Perez, J. Grimm, et al., An X-ray computed tomography imaging agent based on long-circulating bismuth sulphide NPs. Nature Materials, 2006, 5: 118-122.

[50]

C.J. Jia, F. Schüth, Colloidal metal NPs as a component of designed catalyst. Physical Chemistry Chemical Physics, 2011, 13: 2457-2487.

[51]

M.A. Faramarzi, A. Sadighi, Insights into biogenic and chemical production of inorganic nanomaterials and nanostructures. Advances in Colloid and Interface Science, 2013, 189: 1-20.

[52]

L.T. Lu, L.D. Tung, I. Robinson, et al., Size and shape control for water-soluble magnetic cobalt NPs using polymer ligands. Journal of Materials Chemistry, 2008, 18: 2453-2458.

[53]

M. Brust, M. Walker, D. Bethell, et al., Synthesis of thiol-derivatised gold NPs in a two-phase Liquid–Liquid system. Journal of the Chemical Society, Chemical Communications, 1994, 801-802.

[54]

N.T.K. Thanh, L.A.W. Green, Functionalisation of NPs for biomedical applications. Nano Today, 2010, 5: 213-230.

[55]

N.R. Jana, L. Gearheart, and C.J. Murphy, Wet chemical synthesis of high aspect ratio cylindrical gold nanorods. The Journal of Physical Chemistry B, 2001, 105: 4065-4067.

[56]

S.K. Das, E. Marsili, A green chemical approach for the synthesis of gold NPs: characterization and mechanistic aspect. Reviews in Environmental Science and Biotechnology, 2010, 9: 199-204.

[57]

J. Bain, S.S. Staniland, Bioinspired nanoreactors for the biomineralisation of metallic-based NPs for nanomedicine. Physical Chemistry Chemical Physics, 2015, 17: 15508-15521.

[58]

V. Bansal, A. Bharde, R. Ramanathan, et al., Inorganic materials using 'unusual' microorganisms. Advances in Colloid and Interface Science, 2012, 179: 150-168.

[59]

A. Prakash, S. Sharma, N. Ahmad, et al., Bacteria mediated extracellular synthesis of metallic NPs. International Research Journal of Biotechnology, 2010, 1: 71-79.

[60]

M. Ashokkumar, Microbial synthesis of silver NPs by Bacillus sp. Journal of Nanoparticle Research, 2009, 11: 1811-1815.

[61]

N. Krumov, S. Oder, I.P. Nochta, et al., Accumulation of CdS NPs by yeasts in a fed-batch bioprocess. Journal of Biotechnology, 2007, 132: 481-487.

[62]

D.H. Nies, Microbial heavy-metal resistance. Applied Microbiology and Biotechnology, 1999, 51: 730-750.

[63]

D. Jaysankar, N. Ramaiah, and L.Vardanyan, Detoxification of toxic heavy metals by marine bacteria highly resistant to mercury. Marine Biotechnology, 2008, 10: 471-477.

[64]

L. Levina, V. Sukhovatkin, S. Musikhin, et al., Efficient infrared-emitting PbS quantum dots grown on DNA and stable in aqueous solution and blood plasma. Advanced Materials, 2005, 15: 1854-1857.

[65]

A. Kumar, B. Singh, Synthesis and photophysics of red emitting RNA templated PbSe nanostructures. Chemical Communications, 2011, 47: 4144-4146.

[66]

S.K. Srivastava, R. Yamada, C. Ogino, et al., Biogenic synthesis and characterization of gold NPs by Escherichia coli K12 and its heterogeneous catalysis in degradation of 4-nitrophenol. Nanoscale Research Letters, 2013, 8: 70.

[67]

K. Sokolov, M. Follen, J. Aaron, et al., Real-time vital optical imaging of precancer using anti-epidermal growth factor receptor antibodies conjugated to gold NPs. Cancer Research, 2003, 63: 1999-2004.

[68]

I.H. El-Sayed, X. Huang, and M.A. El-Sayed, Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold NPs in cancer diagnostics: applications in oral cancer. Nano Letter, 2005, 5: 829-834.

[69]

I.H. El-Sayed, X. Huang, and M.A. El-Sayed, Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold NPs. Cancer Letter, 2006, 28: 129-135.

[70]

Y. Wang, Y. Liu, H. Luehmann, et al., Evaluating the pharmacokinetics and in vivo cancer targeting capability of Au nanocages by positron emission tomography imaging. ACS Nano, 2012, 24: 5880-5888.

[71]

M. Everts, V. Saini, J.L. Leddon, et al., Covalently linked Au NPs to a viral vector: potential for combined photothermal and gene cancer therapy. Nano Letter, 2006, 6: 587-591.

[72]

M.Y. Lan, Y.B. Hsu, C.H. Hsu, et al., Induction of apoptosis by high-dose gold NPs in nasopharyngeal carcinoma cells. Auris Nasus Larynx, 2013, 40: 563-568.

[73]

M. Benito, V. Martín, M.D. Blanco, et al., Cooperative effect of 5-aminolevulinic acid and gold NPs for photodynamic therapy of cancer. Journal of Pharmaceutical Sciences, 2013, 102: 2760-2769.

[74]

P. Karuppaiya, E. Satheeshkumar, W.T. Chao, et al., Anti-metastatic activity of biologically synthesized gold NPs on human fibrosarcoma cell line HT-1080. Colloids and Surfaces B: Biointerfaces, 2013, 110: 163-170.

[75]

E. Amato, A. Italiano, S. Leotta, et al., Monte Carlo study of the dose enhancement effect of gold NPs during X-ray therapies and evaluation of the anti-angiogenic effect on tumour capillary vessels. Journal of X-Ray Science and Technology, 2013, 21: 237-247.

[76]

J. Liu, W. Zhang, H. Zhang, et al., A multifunctional nanoprobe based on Au–Fe3O4 NPs for multimodal and ultrasensitive detection of cancer cells. Chemical Communications (Camb), 2013, 49: 4938-4940.

[77]

R. Govender, A. Phulukdaree, R.M. Gengan, et al., Silver NPs of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line. Journal of Nanobiotechnology, 2013, 11: 5.

[78]

I. Sur, M. Altunbek, M. Kahraman, et al., The influence of the surface chemistry of silver NPs on cell death. Nanotechnology, 2012, 21: 375102.

[79]

R. Lu, D. Yang, D. Cui, et al., Egg white-mediated green synthesis of silver NPs with excellent biocompatibility and enhanced radiation effects on cancer cells. International Journal of Nanomedicine, 2012, 7: 2101-2107.

[80]

L.A. Austin, B. Kang, C.W. Yen, et al., Plasmonic imaging of human oral cancer cell communities during programmed cell death by nuclear-targeting silver NPs. Journal of the American Chemical Society, 2011, 9: 17594-17597.

[81]

S.C. Boca, M. Potara, A.M. Gabudean, et al., Chitosan-coated triangular silver NPs as a novel class of biocompatible, highly effective photothermal transducers for in vitro cancer cell therapy. Cancer Letter, 2011, 311: 131-140.

[82]

L. Liu, F. Ni, J. Zhang, et al., Silver nanocrystals sensitize magnetic-nanoparticle-mediated thermo-induced killing of cancer cells. Acta Biochimica et Biophysica Sinica, 2011, 43: 316-323.

[83]

P. Sanpui, A. Chattopadhyay, S.S. Ghosh, Induction of apoptosis in cancer cells at low silver nanoparticle concentrations using chitosan nanocarrier. ACS Applied Materials & Interfaces, 2011, 2: 218-228.

[84]

H. Wang, Y. Zhang, H. Yu, et al., Label-free electrochemical immunosensor for prostate-specific antigen based on silver hybridized mesoporous silica NPs. Analytical Biochemistry, 2013, 434: 123-127.

[85]

E. Locatelli, F. Broggi, J. Ponti, et al., Lipophilic silver NPs and their polymeric entrapment into targeted-PEG-based micelles for the treatment of glioblastoma. Advanced Healthcare Materials, 2012, 1: 342-347.

[86]

S.N. Ostad, S. Dehnad, Z.E. Nazari, et al., Cytotoxic activities of silver NPs and silver ions in parent and tamoxifen-resistant T47D human breast cancer cells and their combination effects with tamoxifen against resistant cells. Avicenna Journal of Medical Biotechnology, 2010, 4: 187-196.

[87]

V. Economopoulos, Y. Chen, C. McFadden, et al., MRI detection of nonproliferative tumor cells in lymph node metastases using iron oxide particles in a mouse model of breast cancer. Translational Oncology, 2013, 6: 347-354.

[88]

C.D. Burford, K.D. Bhattacharyya, N. Boriraksantikul, et al,. Nanoparticle mediated thermal ablation of breast cancer cells using a nanosecond pulsed electric field. IEEE Transactions on NanoBioscience, 2013, 2: 112-118.

[89]

S.A. Meenach, J.M. Shapiro, J.Z. Hilt, et al., Characterization of PEG-iron oxide hydrogel nanocomposites for dual hyperthermia and paclitaxel delivery. Journal of Biomaterials Science, Polymer Edition, 2013, 24: 1112-1126.

[90]

X. Wang, B. Chen, X. Yang,et al., Functionalized superparamagnetic NPs for highly-efficient gene delivery. Journal of Nanoscience and Nanotechnology, 2013, 2: 746-750.

[91]

L. Zhao, M. Huo, J. Liu, et al., In vitro investigation on the magnetic thermochemotherapy mediated by magnetic NPs combined with methotrexate for breast cancer treatment. Journal of Nanoscience and Nanotechnology, 2013, 2: 741-745.

[92]

K. Hayashi, M. Nakamura, W. Sakamoto, et al., Superparamagnetic nanoparticle clusters for cancer theranostics combining magnetic resonance imaging and hyperthermia treatment. Theranostics, 2013, 3: 366-376.

[93]

S. Mahdihassan, Cinnabar-gold as the best alchemical drug of longevity, called Makaradhwaja in India. American Journal of Chinese Medicine, 1985, 13: 93-108.

[94]

S.P. Fricker, R.G. Buckley, Comparison of two colorimetric assays as cytotoxicity endpoints for an in vitro screen for antitumour agents. Anticancer Research, 1996, 16: 3755-3760.

[95]

D.T. Felson, J.J. Anderson, and R.F. Meenan, The comparative efficacy and toxicity of second-line drugs in rheumatoid arthritis. Results of two metaanalyses. Arthritis & Rheumatology, 1990, 33: 1449-1461.

[96]

R. Bhattacharya, P. Mukherjee, Biological properties of "naked" metal NPs. Advanced Drug Delivery Reviews, 2008, 60: 1289-1306.

[97]

D. Cabuzu, A. Cirja, R. Puiu et al., Biomedical applications of gold NPs. Current Topics in Medicinal Chemistry, 2015, 16: 1605-1613.

[98]

Y. Levy, J.N. Onuchic, Mechanisms of protein assembly: lessons from minimalist models. Accounts of Chemical Research, 2006, 39: 135-142.

[99]

A. Hamilton, G. Hortobagyi, Chemotherapy: what progress in the last 5 years? Journal of Clinical Oncology, 2005, 23: 1760-1775.

[100]

I.H. El-Sayed, X. Huang, and M.A. El-Sayed, Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold NPs. Cancer Letter, 2006, 239: 129-135.

[101]

C.R. Patra, R. Bhattacharya, D. Mukhopadhyay, et al., Fabrication of gold NPs for targeted therapy in pancreatic cancer. Advanced Drug Delivery Reviews, 2010, 62: 346-361.

[102]

C.J. Gannon, C.R. Patra, R. Bhattacharya, et al., Intracellular gold NPs enhance non-invasive radiofrequency thermal destruction of human gastrointestinal cancer cells. Journal of Nanobiotechnology, 2008, 6: 2.

[103]

N. Chattopadhyay, Z. Cai, Y.L. Kwon, et al., Molecularly targeted gold NPs enhance the radiation response of breast cancer cells and tumor xenografts to X-radiation. Breast Cancer Research and Treatment, 2013, 137: 81-91.

[104]

P.K. Jain, I.H. El-Sayed, and M.A. El-Sayed, Au NPs target cancer. Nano Today, 2007, 2: 18-29.

[105]

D.P. O'Neal, L.R. Hirsch, N.J. Halas, et al., Photo-thermal tumor ablation in mice using near infrared-absorbing NPs. Cancer Letter, 2004, 209: 171-176.

[106]

I.H. El-Sayed, X. Huang, and M.A. El-Sayed, Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold NPs in cancer diagnostics: applications in oral cancer. Nano Letter, 2005, 5: 829-834.

[107]

Y. Wang, X. Xie, X. Wang, et al., Photoacoustic tomography of a nanoshell contrast agent in the in vivo rat brain. Nano Letter, 2004, 4: 1689-1692.

[108]

W. Faulk, G. Taylor, An immunocolloid method for the electron microscope. Immunochemistry, 1971, 8: 1081-1083.

[109]

C. You, C. Han, X. Wang, et al., The progress of silver NPs in the antibacterial mechanism, clinical application and cytotoxicity. Molecular Biology Reports, 2012, 39: 9193-9201.

[110]

M. Ahamed , M.S. Alsalhi, and M.K. Siddiqui, Silver nanoparticle applications and human health. Clinica Chimica Acta, 2010, 14: 1841-1848.

[111]

H.J. Johnston, G. Hutchison, F.M. Christensen, et al., A review of the in vivo and in vitro toxicity of silver and gold particulates: particle attributes and biological mechanisms responsible for the observed toxicity. Critical Reviews in Toxicology, 2010, 40: 328-346.

[112]

S.C.G. K. Daniel, R. Kumar, V. Sathish, et al., Green synthesis (Ocimum tenuiflorum) of silver NPs and toxicity studies in zebra fish (Danio rerio) model. International Journal of Nanoscience and Nanotechnology, 2011, 2: 103-117.

[113]

P. Bourrinet, H.H. Bengele, B. Bonnemain, et al., Preclinical safety and pharmacokinetic profile of ferumoxtran-10, an ultrasmall superparamagnetic iron oxide magnetic resonance contrast agent. Investigative Radiology, 2006, 41: 313-324.

[114]

R. Vaidyanathan, K. Kalishwaralal, S. Gopalram, et al., Nanosilver-the burgeoning therapeutic molecule and its green synthesis. Biotechnology Advances, 2009, 27: 924-937.

[115]

M. Jeyaraj, G. Sathishkumar, G. Sivanandhan, et al., A. Biogenic silver NPs for cancer treatment: an experimental report. Colloids and Surfaces B: Biointerfaces, 2013, 106: 86-92.

[116]

O.V. Melnikov, O.Y. Gorbenko, M.N. Markelova, et al,. Ag-doped manganite NPs: new materials for temperature-controlled medical hyperthermia. Journal of Biomedical Materials Research A, 2009, 91: 1048-1055.

[117]

P.V.A. Rani, G.L.K. Mun, M.P. Hande, et al., Cytotoxicity and genotoxicity of silver NPs in human cells. ACS Nano, 2009, 3: 279-290.

[118]

T. Xia, M. Kovochich, J. Brant, et al., Comparison of the abilities of ambient and manufactured NPs to induce cellular toxicity according to an oxidative stress paradigm. Nano Letter, 2006, 6: 1794-1807.

[119]

M.F. Caroline, J.S. Graeme, G.F.G.T. George, et al., Effects of anti-epilepticdrugs on glutamine synthetase activity in mouse brain. British Journal of Pharmacology, 1999, 126: 1634-1638.

[120]

R. Ranganathan, S. Madanmohan, A. Kesavan, et al., Nanomedicine: towards development of patient-friendly drug-delivery systems for oncological applications. International Journal of Nanomedicine, 2012, 7: 1043-1060.

[121]

Q. Wu, H. Cao, Q. Luan, et al., Biomolecule-assisted synthesis of water soluble silver NPs and their biomedical applications. Inorganic Chemistry, 2008, 47: 5882-5888.

[122]

A.N. Ananth, S.C.G. K. Daniel, T.A. Sironmani, et al., PVA and BSA stabilized silver NPs based surface—enhanced plasmon resonance probes for protein detection. Colloid Surface B, 2011, 85: 138-144.

[123]

G.F. Goya, T.S. Berquo, F.C. Fonseca, Static and dynamic magnetic properties of spherical magnetite NPs. Journal of Applied Physics, 2003, 94: 3520-3527.

[124]

F. Yang, C. Jin, S. Subedi, et al., Emerging inorganic nanomaterials for pancreatic cancer diagnosis and treatment. Cancer Treatment Reviews, 2012, 38: 566-579.

[125]

A.K. Gupta, M. Gupta, Synthesis and surface engineering of iron oxide NPs for biomedical applications. Biomaterials, 2005, 26: 3995-4021.

[126]

D.R. Wahajuddin, S. Arora, Superparamagnetic iron oxide NPs: magnetic nanoplatforms as drug carriers. International Journal of Nanomedicine, 2012, 7: 3445-3471.

[127]

K. Pusic, Z. Aguilar, J. McLoughlin, et al., Iron oxide NPs as a clinically acceptable delivery platform for a recombinant blood-stage human malaria vaccine. FASEB Journal, 2013, 27: 1153-1166.

[128]

A. Mandal, S. Sekar, M. Kanagavel, et al., Collagen based magnetic nanobiocomposite as MRI contrast agent and for targeted delivery in cancer therapy. Biochimica et Biophysica Acta, 2013, 1830: 4628-4633.

[129]

D. Bhattacharya, M. Das, D. Mishra, et al., Folate receptor targeted, carboxymethyl chitosan functionalized iron oxide NPs: a novel ultradispersed nanoconjugates for bimodal imaging. Nanoscale, 2011, 3: 1653-1662.

[130]

M. Chamundeeswari, T.P. Sastry, B.S. Lakhsmi, et al., Iron NPs from animal blood for cellular imaging and targeted delivery for cancer treatment. Biochimica et Biophysica Acta, 2013, 1830: 3005-3010.

[131]

A.M. El-Toni, M.A. Ibrahim, J.P. Labis, et al., Optimization of synthesis parameters for mesoporous shell formation on magnetic nanocores and their application as nanocarriers for docetaxel cancer drug. International Journal of Molecular Sciences, 2013, 30: 11496-11509.

[132]

H. Zhao, Z. Li, B. Yang et al., Synthesis of dual-functional targeting probes for cancer theranostics based on iron oxide NPs coated by centipede-like polymer connected with pH-responsive anticancer drug. Journal of Biomaterials Science, Polymer Edition, 2015, 7: 1-30.

[133]

A.H. Lu, E.L. Salabas, and F. Schuth, Magnetic NPs: synthesis, protection, functionalization, and application. Angewandte Chemie International Edition in English, 2007, 46: 1222-1244.

[134]

O.S. Nielsen, M. Horsman, and J. Overgaard, A future for hyperthermia in cancer treatment? European Journal of Cancer, 2001, 37: 1587-1589.

[135]

A. Ito, M. Shinkai, H. Honda, et al., Heat-inducible TNF-alpha gene therapy combined with hyperthermia using magnetic NPs as a novel tumor-targeted therapy. Cancer Gene Therapy, 2001, 8: 649-654.

[136]

H.L.R. Luccioni, M.L. Esteves, J.M. Vega, et al., Enhanced reduction in cell viability by hyperthermia induced by magnetic NPs. International Journal of Nanomedicine, 2011, 6: 373-380.

[137]

C.A. Quinto, P. Mohindra, S. Tong et al., Multifunctional superparamagnetic iron oxide NPs for combined chemotherapy and hyperthermia cancer treatment. Nanoscale, 2015, 7: 12728-12736.

[138]

A. Liberman, H.P. Martinez, C.N. Ta, et al., Hollow silica and silica-boron nano/microparticles for contrast-enhanced ultrasound to detect small tumors. Biomaterials, 2012, 33: 5124-5129.

[139]

A.V. Efremenko, A.A. Ignatova, A.A. Borsheva, et al., Cobalt bis(dicarbollide) versus closo-dodecaborate in boronated chlorin e(6) conjugates: implications for photodynamic and boron-neutron capture therapy. Photochemical & Photobiological Sciences, 2012, 11: 645-652.

[140]

C.H. Lai, Y.C. Lin, F.I. Chou, et al ., Design of multivalent galactosyl carborane as a targeting specific agent for potential application to boron neutron capture therapy. Chemical Communications (Camb), 2012, 48: 612-614.

[141]

C. Hong, C. Lee, In vitro cell tests of pancreatic malignant tumor cells by photothermotherapy based on DMSO porous silicon colloids. Lasers in Medical Science, 2013, 20: 221-223.

[142]

E. Secret, M. Maynadier, A. Gallud, et al., Anionic porphyrin-grafted porous silicon NPs for photodynamic therapy. Chemical Communications (Camb), 2013, 49: 4202-4204.

[143]

C. Qian, Y. Wang, Y. Chen, et al., Suppression of pancreatic tumor growth by targeted arsenic delivery with anti-CD44v6 single chain antibody conjugated NPs. Biomaterials, 2013, 34: 6175-6184.

[144]

R.W. Ahn, F. Chen, H. Chen, et al., A novel nanoparticulate formulation of arsenic trioxide with enhanced therapeutic efficacy in a murine model of breast cancer. Clinical Cancer Research, 2010, 16: 3607-3617.

[145]

H. Chen, R. Ahn, J. Van den Bossche, et al., Folate-mediated intracellular drug delivery increases the anticancer efficacy of nanoparticulate formulation of arsenic trioxide. Molecular Cancer Therapeutics, 2009, 8: 1955-1963.

[146]

Z.Y. Wang, J. Song, D.S. Zhang, Nanosized As2O3/Fe2O3 complexes combined with magnetic fluid hyperthermia selectively target liver cancer cells. World Journal of Gastroenterology, 2009, 15: 2995-3002.

[147]

J. Li, F. Jiang, B. Yang, et al., Topological insulator bismuth selenide as a theranostic platform for simultaneous cancer imaging and therapy. Scientific Reports, 2013, 17: 1998.

[148]

M. Hossain, Y. Luo, Z. Sun, et al., X-ray enabled detection and eradication of circulating tumor cells with NPs. Biosensors and Bioelectronics, 2012, 38: 348-354.

[149]

J.M. Kinsella, R.E. Jimenez, P.P. Karmali, et al., X-ray computed tomography imaging of breast cancer by using targeted peptide-labeled bismuth sulfide NPs. Angewandte Chemie International Edition in English, 2011, 51: 12308-12311.

[150]

M. Chu, X. Pan, D. Zhang, et al., The therapeutic efficacy of CdTe and CdSe quantum dots for photothermal cancer therapy. Biomaterials, 2012, 33: 7071-7073.

[151]

J. Ruan, H. Song, Q. Qian, et al., HER2 monoclonal antibody conjugated RNase-A-associated CdTe quantum dots for targeted imaging and therapy of gastric cancer. Biomaterials, 2012, 33: 7093-7102.

[152]

Y. Wang, Y. Zhang, Z. Du, et al., Detection of micrometastases in lung cancer with magnetic NPs and quantum dots. International Journal of Nanomedicine, 2012, 7: 2315-2324.

[153]

A. Sharma, C.M. Pandey, G. Sumana, et al., Chitosan encapsulated quantum dots platform for leukemia detection. Biosensors and Bioelectronics, 2012, 38: 107-113.

[154]

C.Y. Tian, W.W. Zhao, J. Wang, et al., Amplified quenching of electrochemiluminescence from CdS sensitized TiO2 nanotubes by CdTe-carbon nanotube composite for detection of prostate protein antigen in serum. Analyst, 2012, 137: 3070-3075.

[155]

H.J. Hu, S.G. Li, J. Liu, et al., Detection of the expression of HER2 using CdTe/ZnSe core/shell quantum dots as fluorescence probe in breast cancer cells. Zhonghua Bing Li Xue Za Zhi, 2011, 40: 484-486.

[156]

V.A. Gérard, C.M. Maguire, D. Bazou, et al., Folic acid modified gelatine coated quantum dots as potential reagents for in vitro cancer diagnostics. Nanobiotechnology, 2011, 9: 50.

[157]

S. Steponkiene, S. Kavaliauskiene, R. Purviniene, et al., Quantum dots affect expression of CD133 surface antigen in melanoma cells. International Journal of Nanomedicine, 2011, 6: 2437-2444.

[158]

V. Morosini, T. Bastogne, C. Frochot, et al., Quantum dot-folic acid conjugates as potential photosensitizers in photodynamic therapy of cancer. Photochemical & Photobiological Sciences, 2011, 10: 842-851.

[159]

A. Rakovich, T. Rakovich, V. Kelly, et al., Photosensitizer methylene blue-semiconductor nanocrystals hybrid system for photodynamic therapy. Journal of Nanoscience and Nanotechnology, 2010, 10: 2656-2662.

[160]
I.V. Sukhorukova, I.Y. Zhitnyak, A.M. Kovalskii, et al., Boron nitride NPs with a petal-like surface as anticancer drug-delivery systems. ACS Applied Material Interfaces, 2015, 31. PMID: 26192448.
[161]

M.J. Luderer, P. de la Puente, and A.K. Azab, Advancements in tumor targeting strategies for boron neutron capture therapy. Pharmaceutical Research, 2015, 32: 2824-2836.

[162]

C.F. Wang, M.P. Sarparanta, E.M. Mäkilä, et al., Multifunctional porous silicon NPs for cancer theranostics. Biomaterials, 2015, 48: 108-118.

[163]

C. Printz, Arsenic nanoparticle holds promise in blocking aggressive breast cancer. Cancer, 2010, 116: 5567.

[164]

R.L. Cunha, I.E. Gouvea, and L. Juliano, A glimpse on biological activities of tellurium compounds. Anais da Academia Brasileira de Ciências, 2009, 81: 393-407.

[165]

D.B. Louria, M.M. Joselow, and A.A. Browder, The human toxicity of certain trace elements. Annals of Internal Medicine, 1972, 76: 307-319.

[166]

A. Sadaf, B. Zeshan, Z. Wang, et al., Toxicity evaluation of hydrophilic CdTe quantum dots and CdTe@ SiO2 NPs in mice. Journal of Nanoscience and Nanotechnology, 2012, 11: 8287-8292.

[167]

F. Liu, Y. Zhang, S. Ge, et al., Magnetic graphene nanosheets based electrochemiluminescence immunoassay of cancer biomarker using CdTe quantum dots coated silica nanospheres as labels. Talanta, 2012, 99: 512-519.

[168]

S.B. Rizvi, L. Yildirimer, S. Ghaderi, et al., A novel POSS-coated quantum dot for biological application. International Journal of Nanomedicine, 2012, 7: 3915-3927.

[169]

B. Sredni, Immunomodulating tellurium compounds as anti-cancer agents. Seminars in Cancer Biology, 2012, 22: 60-69.

[170]

B. Sredni, A. Shani, R. Catane, et al., Predominance of TH1 response in tumor-bearing mice and cancer patients treated with AS101. Journal of the National Cancer Institute, 1996, 88: 1276-1284.

[171]

B. Sredni, R. Gal, I.J. Cohen, et al., Hair growth induction by the Tellurium immunomodulator AS101: association with delayed terminal differentiation of follicular keratinocytes and ras-dependent up-regulation of KGF expression. FASEB Journal, 2004, 18: 400-402.

[172]

S.L. Goodman, M. Picard, Integrins as therapeutic targets. Trends in Pharmacological Sciences, 2012, 33: 405-412.

[173]

B.J. Allen, S. Rizvi, Y. Li, et al., In vitro and preclinical targeted alpha therapy for melanoma, breast, prostate and colorectal cancers. Critical Reviews in Oncology/Hematology, 2001, 39: 139-146.

[174]

M. Hossain, M. Su, Nanoparticle location and material dependent dose enhancement in X-ray radiation therapy. Journal of Physical Chemistry C: Nanomaterials and Interfaces, 2012, 116: 23047-23052.

[175]

P. Nazari, R.D. Bazaz, M.R. Mofid, et al., The antimicrobial effects and metabolomic footprinting of carboxyl-capped bismuth NPs against Helicobacter pylori. Applied Biochemistry and Biotechnology, 2014, 172: 570-579.

[176]

H. Zeng, G.F. Combs Jr., Selenium as an anticancer nutrient: roles in cell proliferation and tumor cell invasion. Journal of Nutritional Biochemistry, 2008, 19: 1-7.

[177]

G. Schrauzer, Selenium yeast: composition, quality, analysis, and safety. Pure Applied Chemistry, 2006, 78: 105-109.

[178]

G. Dennert, M. Zwahlen, M. Brinkman, et al., Selenium for preventing cancer. Cochrane Database of Systematic Reviews, 2011, 11 (5): CD005195.

[179]

H. Tapiero, D.M. Townsend, K.D. Tew, The antioxidant role of selenium and seleno-compounds. Biomedicine & Pharmacotherapy, 2003, 57: 134-144.

[180]

C.D. Davis, P.A. Tsuji, and J.A. Milner, Selenoproteins and cancer prevention. Annual Review of Nutrition, 2012, 21: 73-95.

[181]

P.A. Sheridan, N. Zhong, B.A. Carlson, et al., Decreased selenoprotein expression alters the immune response during influenza virus infection in mice. Journal of Nutrition, 2007, 137: 1466-1471.

[182]

L.B. Yang, Y.H. Shen, A.J. Xie, et al., Synthesis of Se NPs by using TSA ion and its photocatalytic application for decolorization of cango red under UV irradiation. Materials Research Bulletin, 2008, 43: 572-582.

[183]

T. Takahashi, S. Yagi, T. Sagawa, et al., X-ray photoemission study of orthorhombic selenium; a new allotrope of crystalline selenium. Journal of the Physical Society of Japan, 1985, 54: 1018-1022.

[184]

J. Zhang, X. Wang, and T. Xu, Elemental selenium at nano size (Nano-Se) as a potential chemopreventive agent with reduced risk of selenium toxicity: comparison with se-methylselenocysteine in mice. Toxicological Sciences, 2007, 101: 22-31.

[185]

J.S. Zhang, X.Y. Gao, L.D. Zhang, et al., Biological effects of a nano red elemental selenium. Biofactors, 2001, 15: 27-38.

[186]

Y. Zhang, X. Li, Z. Huang, et al., Enhancement of cell permeabilization apoptosis-inducing activity of selenium NPs by ATP surface decoration. Nanomedicine, 2013, 9: 74-84.

[187]

Y. Ren, T. Zhao, G. Mao, et al., Antitumor activity of hyaluronic acid-selenium NPs in Heps tumor mice models. International Journal of Biological Macromolecules, 2013, 57: 57-62.

[188]

C. Ramamurthy, K.S. Sampath, P. Arunkumar, et al., Green synthesis and characterization of selenium NPs and its augmented cytotoxicity with doxorubicin on cancer cells. Bioprocess and Biosystems Engineering, 2013, 3: 1131-1139.

[189]

M.H. Yazdi, M. Mahdavi, E. Kheradmand, et al., The preventive oral supplementation of a selenium nanoparticle-enriched probiotic increases the immune response and lifespan of 4T1 breast cancer bearing mice. Arzneimittelforschung, 2012, 62: 525-531.

[190]

M.H. Yazdi, M. Mahdavi, B. Varastehmoradi, et al., The immunostimulatory effect of biogenic selenium NPs on the 4T1 breast cancer model: an in vivo study. Biological Trace Element Research, 2012, 149: 22-28.

[191]

M.H. Yazdi, M. Mahdavi, N. Setayesh, et al., Selenium nanoparticle-enriched Lactobacillus brevis causes more efficient immune responses in vivo and reduces the liver metastasis in metastatic form of mouse breast cancer. DARU Journal of Pharmaceutical Sciences, 2013, 21(1): 33.

[192]

D. Sun, Y. Liu, Q. Yu, et al., The effects of luminescent ruthenium(Ⅱ) polypyridyl functionalized selenium NPs on bFGF-induced angiogenesis and AKT/ERK signaling. Biomaterials, 2013, 34: 171-80.

[193]

S. Zheng, X. Li, Y. Zhang, et al., PEG-nanolized ultrasmall selenium NPs overcome drug resistance in hepatocellular carcinoma HepG2 cells through induction of mitochondria dysfunction. International Journal of Nanomedicine, 2012, 7: 3939-3949.

[194]

B. Yu, Y. Zhang, W. Zheng, et al., Positive surface charge enhances selective cellular uptake and anticancer efficacy of selenium NPs. Inorganic Chemistry, 2012, 20: 8956-8963.

[195]

W. Liu, X. Li, Y.S. Wong, et al., Selenium NPs as a carrier of 5-fluorouracil to achieve anticancer synergism. ACS Nano, 2012, 6: 6578-6591.

[196]

Y.J. Choi, Y.J. Kim, J.W. Lee, et al., Cyto-/genotoxic effect of CdSe/ZnS quantum dots in human lung adenocarcinoma cells for potential photodynamic UV therapy applications. Journal for Nanoscience and Nanotechnology, 2012, 12: 2160-2168.

[197]

F. Yang, Q. Tang, X. Zhong, et al., Surface decoration by Spirulina polysaccharide enhances the cellular uptake and anticancer efficacy of selenium NPs. International Journal of Nanomedicine, 2012, 7: 835-844.

[198]

X. Gao, H. Zhang, Y. Li, et al., Mn-doped ZnSe d-dots-based α-methylacyl-CoA racemase probe for human prostate cancer cell imaging. Analytical and Bioanalytical Chemistry, 2012, 402: 1871-1877.

[199]

L. Hong, Z. Wang, L. Yuan, et al., Subcellular distribution of CdSe quantum dots (QDs) in breast cancer cells. Journal for Nanoscience and Nanotechnology, 2012, 12: 365-367.

[200]

K.K. Vekariya, J. Kaur, K. Tikoo. ERα signaling imparts chemotherapeutic selectivity to selenium NPs in breast cancer. Nanomedicine, 2012, 8: 1125-1132.

[201]

L. Kong, Q. Yuan, H. Zhu, et al., The suppression of prostate LNCaP cancer cells growth by Selenium NPs through Akt/Mdm2/AR controlled apoptosis. Biomaterials, 2011, 32: 6515-6522.

[202]

H. Luo, F. Wang, Y. Bai, et al., Selenium NPs inhibit the growth of HeLa and MDA-MB-231 cells through induction of S phase arrest. Colloids and Surfaces B: Biointerfaces, 2012, 1: 304-308.

[203]

M. Shakibaie, H. Forootanfar, K.M. Moghaddam, et al., Green synthesis of gold NPs by the marine microalga Tetraselmis suecica. Biotechnology and Applied Biochemistry, 2010, 57: 71-75.

[204]

M.M.S. Dallal, M.H. Yazdi, M. Holakuyee, et al., Lactobacillus casei ssp.casei induced Th1 cytokine profile and natural killer cells activity in invasive ductal carcinoma bearing mice. Iranian Journal of Allergy, Asthma and Immunology, 2012, 11: 183-189.

[205]

M.H. Yazdi, M.M.S. Dallal, Z.M. Hassan, et al., Oral administration of Lactobacillus acidophilus induces IL-12 production in spleen cell culture of BALB/c mice bearing transplanted breast tumour. British Journal of Nutrition, 2010, 104: 227-232.

[206]

E. Faghfuri , M.H. Yazdi, M. Mahdavi et al., Dose-response relationship study of selenium NPs as an immunostimulatory agent in cancer-bearing mice. Archive in Medical Researches, 2015, 46: 31−37.

[207]

S. Chatterjee, S. Kundu, and A. Bhattacharyya, Mechanism of cadmium induced apoptosis in the immunocyte. Toxicology Letter, 2008, 177: 83-89.

[208]

N. Pathak, S. Khandelwal, Role of oxidative stress and apoptosis in cadmium induced thymic atrophy and splenomegaly in mice. Toxicology Letter, 2007, 169: 95-108.

[209]

A.C. Uğuz, M. Naziroğlu, J. Espino, et al., Selenium modulates oxidative stress-induced cell apoptosis in human myeloid HL-60 cells through regulation of calcium release and caspase-3 and -9 activities. Journal of Membrane Biology, 2009, 232: 15-23.

[210]

M. Shakibaie, A.R. Shahverdi, M.A. Faramarzi, et al., Acute and subacute toxicity of novel biogenic selenium NPs in mice. Pharmaceutical Biology, 2013, 51: 58-63.

[211]

C.C. Liang, A.Y. Park, J.L. Guan, In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nature Protocols, 2007, 2: 329-333.

[212]

V. Jamier, L.A. Ba, and C. Jacob, Selenium- and tellurium-containing multifunctional redox agents as biochemical redox modulators with selective cytotoxicity. Chemistry, 2010, 16: 10920-10928.

Nano Biomedicine and Engineering
Pages 246-267
Cite this article:
Yazdi MH, Sepehrizadeh Z, Mahdavi M, et al. Metal, Metalloid, and Oxide Nanoparticles for Therapeutic and Diagnostic Oncology. Nano Biomedicine and Engineering, 2016, 8(4): 246-267. https://doi.org/10.5101/nbe.v8i4.p246-267

393

Views

9

Downloads

18

Crossref

19

Scopus

Altmetrics

Received: 24 September 2016
Accepted: 21 November 2016
Published: 06 December 2016
© 2016 Mohammad Hossein Yazdi, Zargham Sepehrizadeh, Mehdi Mahdavi, Ahmad Reza Shahverdi, and Mohammad Ali Faramarzi.

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Return