Skip to main content
Top
Published in: Current Oncology Reports 2/2010

Open Access 01-03-2010

Nanotechnology in Head and Neck Cancer: The Race Is On

Author: Ivan H. El-Sayed

Published in: Current Oncology Reports | Issue 2/2010

Login to get access

Abstract

Rapid advances in the ability to produce nanoparticles of uniform size, shape, and composition have started a revolution in the sciences. Nano-sized structures herald innovative technology with a wide range of potential therapeutic and diagnostic applications. More than 1000 nanostructures have been reported, many with potential medical applications, such as metallic-, dielectric-, magnetic-, liposomal-, and carbon-based structures. Of these, noble metallic nanoparticles are generating significant interest because of their multifunctional capacity for novel methods of laboratory-based diagnostics, in vivo clinical diagnostic imaging, and therapeutic treatments. This review focuses on recent advances in the applications of nanotechnology in head and neck cancer, with special emphasis on the particularly promising plasmonic gold nanotechnology.
Literature
1.
go back to reference El-Sayed MA: Some interesting properties of metals confined in time and nanometer space of different shapes. Acc Chem Res 2001, 34:257–264.CrossRefPubMed El-Sayed MA: Some interesting properties of metals confined in time and nanometer space of different shapes. Acc Chem Res 2001, 34:257–264.CrossRefPubMed
2.
go back to reference Pridgen EM, Langer R, Farokhzad OC: Biodegradable, polymeric nanoparticle delivery systems for cancer therapy. Nanomed 2007, 2:669–680.CrossRef Pridgen EM, Langer R, Farokhzad OC: Biodegradable, polymeric nanoparticle delivery systems for cancer therapy. Nanomed 2007, 2:669–680.CrossRef
3.
go back to reference Huang X, Jain PK, El-Sayed IH, El-Sayed MA: Gold nanoparticles: interesting optical properties and recent applications in cancer diagnostics and therapy. Nanomed 2007, 2:681–693.CrossRef Huang X, Jain PK, El-Sayed IH, El-Sayed MA: Gold nanoparticles: interesting optical properties and recent applications in cancer diagnostics and therapy. Nanomed 2007, 2:681–693.CrossRef
4.
go back to reference Huang X, El-Sayed IH, Qian W, El-Sayed MA: Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J Am Chem Soc 2006, 128:2115–2120.CrossRefPubMed Huang X, El-Sayed IH, Qian W, El-Sayed MA: Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J Am Chem Soc 2006, 128:2115–2120.CrossRefPubMed
5.
go back to reference El-Sayed IH, Huang X, El-Sayed MA: Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett 2006, 239:129–135.CrossRefPubMed El-Sayed IH, Huang X, El-Sayed MA: Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett 2006, 239:129–135.CrossRefPubMed
6.
go back to reference •• Jain PK, El-Sayed IH, El-Sayed MA: Au nanoparticles target cancer. NanoToday 2007, 2:18–29. This article presents a thorough review of the optical properties of gold nanoparticles useful for imaging and treatment of cancer. •• Jain PK, El-Sayed IH, El-Sayed MA: Au nanoparticles target cancer. NanoToday 2007, 2:18–29. This article presents a thorough review of the optical properties of gold nanoparticles useful for imaging and treatment of cancer.
7.
go back to reference •Dickerson EB, Dreaden EC, Huang X, et al.: Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett 2008, 269:57–66.CrossRefPubMed •Dickerson EB, Dreaden EC, Huang X, et al.: Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett 2008, 269:57–66.CrossRefPubMed
8.
go back to reference Paciotti GF, Kingston DG, Tamarkin L: Colloidal gold nanoparticles: a novel nanoparticle platform for developing multifunctional tumor-targeted drug delivery vectors. Drug Dev Res 2006, 67:47–54.CrossRef Paciotti GF, Kingston DG, Tamarkin L: Colloidal gold nanoparticles: a novel nanoparticle platform for developing multifunctional tumor-targeted drug delivery vectors. Drug Dev Res 2006, 67:47–54.CrossRef
9.
go back to reference Niidome T, Yamagata M, Okamoto Y, et al.: PEG-modified gold nanorods with a stealth character for in vivo applications. J Control Release 2006, 114:343–347CrossRefPubMed Niidome T, Yamagata M, Okamoto Y, et al.: PEG-modified gold nanorods with a stealth character for in vivo applications. J Control Release 2006, 114:343–347CrossRefPubMed
10.
go back to reference Paciotti GF, Myer L, Weinreich D, et al.: Colloidal gold: a novel nanoparticle vector for tumor directed drug delivery. Drug Deliv 2004, 11:169–183.CrossRefPubMed Paciotti GF, Myer L, Weinreich D, et al.: Colloidal gold: a novel nanoparticle vector for tumor directed drug delivery. Drug Deliv 2004, 11:169–183.CrossRefPubMed
11.
go back to reference Hainfeld JF, Slatkin DN, Smilowitz HM: The use of gold nanoparticles to enhance radiotherapy in mice. Phys Med Biol 2004, 49:N309–N315.CrossRefPubMed Hainfeld JF, Slatkin DN, Smilowitz HM: The use of gold nanoparticles to enhance radiotherapy in mice. Phys Med Biol 2004, 49:N309–N315.CrossRefPubMed
14.
16.
go back to reference Myers EN, Simental AA: Cancer of the oral cavity. In Cancer of the Head and Neck. Edited by Myers EN, Suen JY, Myers JN, Hanna EY. Philadelphia, PA: Saunders; 2003:279–332. Myers EN, Simental AA: Cancer of the oral cavity. In Cancer of the Head and Neck. Edited by Myers EN, Suen JY, Myers JN, Hanna EY. Philadelphia, PA: Saunders; 2003:279–332.
17.
go back to reference Myers EN, Suen JY: Perspectives in head and neck cancer. In Cancer of the Head and Neck. Edited by Myers EN, Suen JY, Myers JN, Hanna EY. Philadelphia, PA: Saunders; 2003:1–5. Myers EN, Suen JY: Perspectives in head and neck cancer. In Cancer of the Head and Neck. Edited by Myers EN, Suen JY, Myers JN, Hanna EY. Philadelphia, PA: Saunders; 2003:1–5.
18.
go back to reference Bonner JA, Harari PM, Giralt J, et al.: Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006, 354:567–578.CrossRefPubMed Bonner JA, Harari PM, Giralt J, et al.: Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006, 354:567–578.CrossRefPubMed
19.
go back to reference Lentsch EJ, Myers JN: Pathogenesis and progression of squamous cell carcinoma of the head and neck. In Cancer of the Head and Neck. Edited by Myers EN, Suen JY, Myers JN, Hanna EY. Philadelphia, PA: Saunders; 2003:5–28. Lentsch EJ, Myers JN: Pathogenesis and progression of squamous cell carcinoma of the head and neck. In Cancer of the Head and Neck. Edited by Myers EN, Suen JY, Myers JN, Hanna EY. Philadelphia, PA: Saunders; 2003:5–28.
20.
go back to reference Iyer AK, Khaled G, Fang J, Maeda H: Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today 2006, 11:812–818.CrossRefPubMed Iyer AK, Khaled G, Fang J, Maeda H: Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today 2006, 11:812–818.CrossRefPubMed
21.
go back to reference Goel R, Shah N, Visaria R, et al.: Biodistribution of TNF-alpha-coated gold nanoparticles in an in vivo model system. Nanomed 2009, 4:401–410.CrossRef Goel R, Shah N, Visaria R, et al.: Biodistribution of TNF-alpha-coated gold nanoparticles in an in vivo model system. Nanomed 2009, 4:401–410.CrossRef
22.
go back to reference Pirollo KF, Chang EF: Does a targeting ligand influence nanoparticle tumor localization or uptake? Trends Biotechnol 2008, 26:552–558.CrossRefPubMed Pirollo KF, Chang EF: Does a targeting ligand influence nanoparticle tumor localization or uptake? Trends Biotechnol 2008, 26:552–558.CrossRefPubMed
23.
go back to reference Yguerabide J, Yguerabide EE: Light-scattering submicroscopic particles as highly fluorescent analogs and their use as tracer labels in clinical and biological applications. I. Theory. Anal Biochem 1998, 262:137–156.CrossRef Yguerabide J, Yguerabide EE: Light-scattering submicroscopic particles as highly fluorescent analogs and their use as tracer labels in clinical and biological applications. I. Theory. Anal Biochem 1998, 262:137–156.CrossRef
24.
go back to reference El-Sayed I, Huang X, Macheret F, Humstoe JO, et al.: Effect of plasmonic gold nanoparticles on benign and malignant cellular autofluorescence: a novel probe for fluorescence based detection of cancer. Technol Cancer Res Treat 2007, 6:403–412.PubMed El-Sayed I, Huang X, Macheret F, Humstoe JO, et al.: Effect of plasmonic gold nanoparticles on benign and malignant cellular autofluorescence: a novel probe for fluorescence based detection of cancer. Technol Cancer Res Treat 2007, 6:403–412.PubMed
25.
go back to reference El-Sayed IH, Huang X, El-Sayed MA: Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics: applications in oral cancer. Nano Lett 2005, 5:829–834.CrossRefPubMed El-Sayed IH, Huang X, El-Sayed MA: Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics: applications in oral cancer. Nano Lett 2005, 5:829–834.CrossRefPubMed
26.
go back to reference Sokolov K, Follen M, Aaron J, et al.: Real-time vital optical imaging of precancer using anti-epidermal growth factor receptor antibodies conjugated to gold nanoparticles. Cancer Res 2004, 63:1999–2004. Sokolov K, Follen M, Aaron J, et al.: Real-time vital optical imaging of precancer using anti-epidermal growth factor receptor antibodies conjugated to gold nanoparticles. Cancer Res 2004, 63:1999–2004.
27.
go back to reference Li PC, Wang CR, Shieh DB, et al.: In vivo photoacoustic molecular imaging with simultaneous multiple selective targeting using antibody-conjugated gold nanorods. Opt Express 2008, 16:18605–18615.CrossRefPubMed Li PC, Wang CR, Shieh DB, et al.: In vivo photoacoustic molecular imaging with simultaneous multiple selective targeting using antibody-conjugated gold nanorods. Opt Express 2008, 16:18605–18615.CrossRefPubMed
28.
go back to reference Zharov VP, Lapotko DO: Photothermal imaging of nanoparticles and cells. IEEE J Selected Topics Quant Elect 2005, 11:733–751.CrossRef Zharov VP, Lapotko DO: Photothermal imaging of nanoparticles and cells. IEEE J Selected Topics Quant Elect 2005, 11:733–751.CrossRef
29.
go back to reference Nikoobakht B, Wang J, El-Sayed MA: Surface-enhanced Raman scattering of molecules adsorbed on gold nanorods: off-surface plasmon resonance condition. Chem Phys Lett 2002, 366:17–23.CrossRef Nikoobakht B, Wang J, El-Sayed MA: Surface-enhanced Raman scattering of molecules adsorbed on gold nanorods: off-surface plasmon resonance condition. Chem Phys Lett 2002, 366:17–23.CrossRef
30.
go back to reference Wang H, Huff TB, Zweifel DA, et al.: In vitro and in vivo two-photon luminescence imaging of single gold nanorods. Proc Natl Acad Sci U S A 2005, 102:15752–15756.CrossRefPubMed Wang H, Huff TB, Zweifel DA, et al.: In vitro and in vivo two-photon luminescence imaging of single gold nanorods. Proc Natl Acad Sci U S A 2005, 102:15752–15756.CrossRefPubMed
31.
go back to reference Song KH, Kim C, Maslov K, Wang LV: Noninvasive in vivo spectroscopic nanorod-contrast photoacoustic mapping of sentinel lymph nodes. Eur J Radiol 2009, 70:227–231.CrossRefPubMed Song KH, Kim C, Maslov K, Wang LV: Noninvasive in vivo spectroscopic nanorod-contrast photoacoustic mapping of sentinel lymph nodes. Eur J Radiol 2009, 70:227–231.CrossRefPubMed
32.
go back to reference Agarwal A, Huang SW, O’Donnell M, et al.: Targeted gold nanorod contrast agent for prostate cancer detection by photoacoustic imaging. J Appl Phys 2007, 102:064701-064701-4. Agarwal A, Huang SW, O’Donnell M, et al.: Targeted gold nanorod contrast agent for prostate cancer detection by photoacoustic imaging. J Appl Phys 2007, 102:064701-064701-4.
33.
go back to reference Niidome T, Akiyama Y, Shimoda K, et al.: In vivo monitoring of intravenously injected gold nanorods using near-infrared light. Small 2008, 4:1001–1007.CrossRefPubMed Niidome T, Akiyama Y, Shimoda K, et al.: In vivo monitoring of intravenously injected gold nanorods using near-infrared light. Small 2008, 4:1001–1007.CrossRefPubMed
34.
go back to reference •• Qian X, Peng XH, Ansari DO, et al.: In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tags. Nat Biotechnol 2008, 26:83–90. This article demonstrates detection of tumor in vivo using SERSs.CrossRefPubMed •• Qian X, Peng XH, Ansari DO, et al.: In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tags. Nat Biotechnol 2008, 26:83–90. This article demonstrates detection of tumor in vivo using SERSs.CrossRefPubMed
35.
go back to reference Aaron J, Nitin N, Travis K, et al.: Plasmon resonance coupling of metal nanoparticles for molecular imaging of carcinogenesis in vivo. J Biomed 2007, 12:034007. Aaron J, Nitin N, Travis K, et al.: Plasmon resonance coupling of metal nanoparticles for molecular imaging of carcinogenesis in vivo. J Biomed 2007, 12:034007.
36.
go back to reference Kneipp J, Kneipp H, Kneippa K: SERS—a single-molecule and nanoscale tool for bioanalytics. Chem Soc Rev 2008, 37:1052–1060.CrossRefPubMed Kneipp J, Kneipp H, Kneippa K: SERS—a single-molecule and nanoscale tool for bioanalytics. Chem Soc Rev 2008, 37:1052–1060.CrossRefPubMed
37.
go back to reference Oyelere AK, Chen B, Huang X, et al.: Peptide conjugated gold nanorods for nuclear targeting. Bioconjugate Chem 2007, 18:1490–1497.CrossRef Oyelere AK, Chen B, Huang X, et al.: Peptide conjugated gold nanorods for nuclear targeting. Bioconjugate Chem 2007, 18:1490–1497.CrossRef
38.
go back to reference Huang X, El-Sayed IH, Qian W, El-Sayed MA: Cancer cells assemble and align Gold nanorods conjugated to antibodies to produce highly enhanced, sharp and polarized surface Raman spectra: a potential cancer diagnostic marker. Nano Lett 2007, 7:1591–1597.CrossRefPubMed Huang X, El-Sayed IH, Qian W, El-Sayed MA: Cancer cells assemble and align Gold nanorods conjugated to antibodies to produce highly enhanced, sharp and polarized surface Raman spectra: a potential cancer diagnostic marker. Nano Lett 2007, 7:1591–1597.CrossRefPubMed
40.
go back to reference Loo C, Lowery A, Halas NJ, et al.: Immunotargeted nanoshells for integrated cancer imaging and therapy. Nano Lett 2005, 5:709–711.CrossRefPubMed Loo C, Lowery A, Halas NJ, et al.: Immunotargeted nanoshells for integrated cancer imaging and therapy. Nano Lett 2005, 5:709–711.CrossRefPubMed
41.
go back to reference Hirsch LR, Stafford RJ, Bankson JA, et al.: Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance. Proc Natl Acad Sci U S A 2003, 100:13549–13554.CrossRefPubMed Hirsch LR, Stafford RJ, Bankson JA, et al.: Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance. Proc Natl Acad Sci U S A 2003, 100:13549–13554.CrossRefPubMed
42.
go back to reference Schwartz JA, Shetty AM, Price RE, et al.: Feasibility study of particle-assisted laser ablation of brain tumors in orthotopic canine model. Cancer Res 2009, 69:1659–1667.CrossRefPubMed Schwartz JA, Shetty AM, Price RE, et al.: Feasibility study of particle-assisted laser ablation of brain tumors in orthotopic canine model. Cancer Res 2009, 69:1659–1667.CrossRefPubMed
43.
go back to reference Jain PK, Lee KS, El-Sayed IH, El-Sayed MA: Calculated absorption and scattering properties of gold nanoparticles of different size, shape, and composition: applications in biological imaging and biomedicine. J Phys Chem B 2006, 110: 7238–7248.CrossRefPubMed Jain PK, Lee KS, El-Sayed IH, El-Sayed MA: Calculated absorption and scattering properties of gold nanoparticles of different size, shape, and composition: applications in biological imaging and biomedicine. J Phys Chem B 2006, 110: 7238–7248.CrossRefPubMed
44.
go back to reference Huang X, Jain PK, El-Sayed IH, El-Sayed MA: Determination of the minimum temperature required for selective photothermal destruction of cancer cells with the use of immunotargeted gold nanoparticles. Photochem Photobiol 2006, 82:412–417.CrossRefPubMed Huang X, Jain PK, El-Sayed IH, El-Sayed MA: Determination of the minimum temperature required for selective photothermal destruction of cancer cells with the use of immunotargeted gold nanoparticles. Photochem Photobiol 2006, 82:412–417.CrossRefPubMed
45.
go back to reference Lapotko D: Therapy with gold nanoparticles and lasers: what really kills the cells? Nanomed 2009, 4:253–256.CrossRef Lapotko D: Therapy with gold nanoparticles and lasers: what really kills the cells? Nanomed 2009, 4:253–256.CrossRef
46.
go back to reference Akiyama Y, Mori T, Katayama Y, Niidome T: The effects of PEG grafting level and injection dose on gold nanorod biodistribution in the tumor-bearing mice. J Control Release 2009, 139:81–84.CrossRefPubMed Akiyama Y, Mori T, Katayama Y, Niidome T: The effects of PEG grafting level and injection dose on gold nanorod biodistribution in the tumor-bearing mice. J Control Release 2009, 139:81–84.CrossRefPubMed
47.
go back to reference Hainfeld JF, Dilmanian FA, Slatkin DN, Smilowitz HM: Radiotherapy enhancement with gold nanoparticles. J Pharm Pharmacol 2008, 60:977–985.CrossRefPubMed Hainfeld JF, Dilmanian FA, Slatkin DN, Smilowitz HM: Radiotherapy enhancement with gold nanoparticles. J Pharm Pharmacol 2008, 60:977–985.CrossRefPubMed
48.
go back to reference Diagaradjane P, Shetty A, Wang JC, et 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 2008, 8:1492–1500.CrossRefPubMed Diagaradjane P, Shetty A, Wang JC, et 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 2008, 8:1492–1500.CrossRefPubMed
49.
go back to reference Peng XH, Qian X, Mao H, et al.: Targeted magnetic iron oxide nanoparticles for tumor imaging and therapy. Int J Nanomedicine 2008, 3:311–321.PubMed Peng XH, Qian X, Mao H, et al.: Targeted magnetic iron oxide nanoparticles for tumor imaging and therapy. Int J Nanomedicine 2008, 3:311–321.PubMed
50.
go back to reference Wang C, Chen J, Talavage T, Irudayaraj J: Gold nanorod/Fe3O4 nanoparticle “nano-pearl-necklaces” for simultaneous targeting, dual-mode imaging, and photothermal ablation of cancer cells. Angew Chem Int Ed Engl 2009, 48:2759–2763.CrossRefPubMed Wang C, Chen J, Talavage T, Irudayaraj J: Gold nanorod/Fe3O4 nanoparticle “nano-pearl-necklaces” for simultaneous targeting, dual-mode imaging, and photothermal ablation of cancer cells. Angew Chem Int Ed Engl 2009, 48:2759–2763.CrossRefPubMed
Metadata
Title
Nanotechnology in Head and Neck Cancer: The Race Is On
Author
Ivan H. El-Sayed
Publication date
01-03-2010
Publisher
Current Science Inc.
Published in
Current Oncology Reports / Issue 2/2010
Print ISSN: 1523-3790
Electronic ISSN: 1534-6269
DOI
https://doi.org/10.1007/s11912-010-0087-2

Other articles of this Issue 2/2010

Current Oncology Reports 2/2010 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine