Advances in nanotechnology research have led to the creation of new generation of contrast agents, therapeutics, and delivery systems. These applications are expected to significantly improve the diagnosis and treatment of a variety of diseases. Two nanotechnologies—semiconductor and metallic nanostructures—are the most advanced in this young field and have been extensively investigated for clinical use. These nanostructures are currently the “model” for the developments of many novel nanostructures. This review describes their chemical design, tunable properties, and utility in medicine. Furthermore, we will describe the current understanding of their toxicity, which could be barriers to their use for human.

Advancements in the synthesis, characterization, and surface modifications of nanoscale structures have provided a foundation for their utility in medical applications.1,4 The unique aspect of these nanostructures is their tunable optical, electronic, magnetic, and biological properties. Due to these properties, the incorporation of nanostructures in detection schemes or the use the nanostructures as contrast agents could improve the specificity and accuracy of current diagnostics.5,9 Furthermore, nanostructures can improve the effectiveness of drugs in the treatment of a variety of diseases when they are used as delivery vehicles because of the increase in therapeutic payload.10,12 In the last fifteen years, researchers have characterized the tunable properties by altering the nanostructure size, shape, and chemical composition and have developed reproducible strategies to make nanostructures of desired properties.13,15 Further, researchers have also developed strategies to render nanostructures biocompatible and capable of being coated with biological molecules.16,19 For example, semiconductor nanocrystals synthesized via organometallic methods contain the hydrophobic ligand tri-n-octylphosphine oxide (TOPO). These nanocrystals can subsequently be made water-soluble by covering the TOPO with the amphiphillic polymer polyacrylic acid grafted with octylamine.20,21 The hydrophylic carboxylic acid groups of the polymer coat render the nanocrystal stable in an aqueous environment. Furthermore, these groups can be used for conjugation to polymers and/or biological molecules such as antibodies and oligonucleotides via the formation of peptide bonds.22,23 See Figure 1  for some surface designs of nanostructures for biomedical applications. Although a select few have advanced to clinical use, many are being examined as in vitro and in vivo diagnostic contrast agents, drug delivery vehicles, and therapeutics. Here, we review two of the oldest and most established nanostructures for biomedical applications: semiconductor quantum dots and gold nanoparticles. We also provide a brief description of nanotoxicity, which could be a potential limitation for their use in biological systems.

Semiconductor nanocrystals are commonly used as probes for labeling cells and tissues owing to their intense, tunable fluorescence.24,28 The origin of this unique physical property is quite interesting. A semiconductor crystal’s band-gap energy becomes strongly size-dependent as its physical dimensions approach the exciton Bohr radius (the natural radius of an electron-hole pair in bulk). This dependence is a result of a phenomenon known as “quantum confinement,” in which the allowed electronic states of the exciton are broadened when its position is restricted by the dimensions of its parent crystal, in a manner analogous to the pedagogical “particle in a box.”29,31 In this size range, semiconductor nanocrystals are considered electronically zero-dimensional and are referred to as quantum dots. The dimension at which quantum confinement becomes significant depends on the exciton Bohr radius in bulk and thus the particular material in question. For the well-studied II–VI direct band-gap semiconducting material cadmium-selenide (CdSe), strong confinement occurs in the range of 1–10 nm.32,34 This results in a shift in fluorescent band-edge emission wavelength from approximately 700 nm down to 450 nm as the crystal size is reduced (Figure 2 ).32,35 

The bright, tunable fluorescence emission of quantum dots has encouraged their use as labels in biomedical applications demanding high sensitivity, spectral resolution, and prolonged imaging. Examples include quantitative in vitro immunoassays5,36,39 and the labeling of fixed cell samples40,41 and tissue specimens.42 Labeling of the membrane receptors of live cells at the single-molecule level has also been achieved.43 This is significant as it allows tracking of membrane receptor diffusion and internalization. In one example, QDs conjugated with epidermal growth factor (EGF) were used to label erB1 receptors to monitor endocytosis and transport along filopodia.44 The erb/HER family of membrane tyrosine kinases are overexpressed in many types of invasive cancer and thought to play a pivotal role in proliferation and differentiation. Other plasma membrane structures have been labeled, including glycine receptors45 and membrane lipids.46 Cytoplasmic structures may also be labeled with QDs for tracking movement, proliferation, and survival in transplanted tissue47 or during embryonic development.48 QDs have also found use as contrast agents in vascular or lymphatic circulation to identify angiogenesis and lymph nodes with a metastatic invasion.49 Finally, QDs have been found to extravasate preferentially in certain tumor types over healthy tissue, with potential significance as an early diagnosis tool and as a guide during surgery.50 

Quasi-spherical nanoscale metallic particles also exhibit interesting optical properties. For instance, light of a wavelength much larger than the diameter of the particle can produce coherent oscillations in the metal’s electron cloud relative to its stationary core. This occurs via the coupling of light in a resonant fashion to free electrons within the metal.51,53 These oscillations are called “surface plasmons,” and lend their name to the effect, which is known as “localized surface plasmon resonance” (LSPR). LSPR results in absorption and scattering of incident radiation at the resonant wavelength, with the strength of the interaction depending on the material. For noble metals, the plasmon resonance condition is satisfied in the visible to near-IR portion of the spectrum, and the coupling is extremely efficient, resulting in a strong extinction. The LSPR wavelength depends as well on the dielectric constant of the surrounding environment and on the presence of inter-particle interactions.54,55 For well-separated gold particles of around 20 nm in an aqueous environment, the resonant condition is satisfied at approximately 520 nm, giving a solution of these particles a distinctive red color (see Figure 3  for details).

Gold nanoparticles are the most commonly studied metallic nanoparticles because of both their ease of synthesis and efficient resonance coupling. The strong dependence of their surface plasmon resonance position and efficiency on the dielectric properties of the surrounding environment offers the potential to create a sensor for adsorption events at the surface. This is a colloidal analogue to the commercialized surface plasmon resonance (SPR) sensor and can be used to monitor antigen-antibody binding kinetics and stoichiometry.54,56 58 

The peak SPR wavelength of gold nanoparticles shifts when they are aggregated due to plasmonic coupling. Single monodisperse gold nanoparticle solutions appear as a red solution but change to blue when they are aggregated. Mirkin and co-workers used this principle to create simple diagnostic systems for detecting proteins and genes.59,62 In their system, an oligonucleotide strand is coated onto the surface of the gold nanoparticle via thiol chemistry. When the gold nanoparticle solution is exposed to a target gene, it hybridizes with the oligonucleotides on the surface of the gold nanoparticles and aggregates them, resulting in a change of solution color from red to blue. The aggregated nanoparticles can be subsequently de-aggregated upon heating. This simple color test for detecting genes (or proteins) can be made in a high-throughput format for the rapid analysis of complex biological samples.63 This principle has also been applied during cell labeling where a 9 nm red-shift in the peak was observed with microabsorption spectroscopy upon gold nanoparticle binding to the EGF receptor in cultured human oral squamous carcinoma cell lines.64 Because of the intense scattering at the LSPR wavelength, gold nanoparticles may be used as labels under confocal reflectance microscopy. This technique was employed, under laser illumination, to image epithelial growth factor receptors (EGFR) in a cervical cancer cell line.65 

One of the most promising areas of application for gold nanoparticles is as an agent for photothermal therapy, or the thermal ablation of tumorigenic cells. Conventional strategies use organic molecules for localized heating, but the absorption cross-section of gold nanoparticles is five orders of magnitude greater than that of the popular agent indocyanine green, making them far more efficient thermal centers. Recently, Hauck et al studied the effect of thermal ablation in several carcinomas using gold nanorods. In addition to observing cell death under laser light illumination, they also showed a strongly synergistic effect when combined with chemotherapeutic agents.66 

Before engineered nanomaterials can be applied in a clinical setting, their potential adverse impact on human health must be evaluated. Much of the initial nanomaterial research overlooked the health risks, in favor of exploring the unique physicochemical properties that enable their use as diagnostic and therapeutic agents. The major toxicological concerns of nanomaterials are (1) the materials is composed of heavy metals with known toxicity and (2) nanomaterials may elicit unique and unpredictable biological responses because of their tunable properties. Further, because of their size, they are accessible to vital cells and organs.49 Within a physiological compartment, many nanomaterials interact with the resident cell populations. This results in either adherence to the surface or internalization by a diverse set of pathways including translocation through the plasma membrane, receptor-mediated endocytosis, and pinocytosis.67,72 Once internalized, particles can remain in endosomes and accumulate within the cell, escape to the cytosol to interact with biomolecules and subcellular organelles, or be recycled to the cell surface.67,68 The eventual localization of the nanomaterial, along with its subsequent cellular interactions, dictates its toxicological effect. For example, a number of studies have shown that nanomaterials are cytotoxic in cultured cell models. For example, CdSe quantum dots were found to be toxic to primary rat hepatocytes73; carbon nanotubes, nanofibers, and nanoparticles were toxic to lung tumor cells74; and iron-containing nanoparticles were toxic to nerve cells.75 However, at the same time, other studies have concluded that certain formulations are non-toxic to cells in culture.48,69 

Recently, researchers have focused on evaluating nanomaterials in animal models, which are more physiologically relevant. In one study, mice injected with long multi-walled carbon nanotubes showed asbestos-like pathogenic effects.76 However, acute and chronic injections of single-walled carbon nanotubes into mice yielded no apparent indication of toxicity, despite localizing in the intestine.77,78 It is worth noting that particles less than 5 nm in size have been observed to be eliminated through renal-filtration, raising the possibility of total-body clearance of nanomaterials and eliminating a great deal of the concern for chronic exposure.79 Similar to the cell data, the experiments from the animals are not conclusive.

As illustrated above, many of the results of these studies are contradictory, with some confirming the toxicity of engineered nanomaterials, while others find no indication of toxicity. This lack of uniformity can be largely attributed to the enormous variety of nanomaterials under study. For example, it is entirely feasible that certain particle formulations are toxic to a particular cell type, while being benign to another. At the same time, a different nanomaterial of a different composition, size, or shape may be toxic to a different set of cells or under a different set of exposure conditions.11 Alternatively, a number of studies have also determined that the reagents used in synthesizing nanoparticles may be the culprits in inducing toxicity, and as a result the nanoparticles could be toxic, not because of the particle itself, but because of the synthetic reagents or chemical by-products.80 This highlights the need for more systematic evaluation of nanoparticle formulations under study as the presence of impurities, adsorbates, and reaction byproducts as well as the stability of a particle system are all important factors. To complicate matters further, there is a lack of standardization in model systems and test assays. An assay may be performed to measure apoptosis, but fail to recognize an arrest in cell proliferation, or a high level of DNA damage or mutation suggesting genotoxicity, leading a researcher to erroneously conclude that the particle is non-toxic.

Nanotechnology is a young research field and enabling technology that still requires development. Despite its youth, nanotechnology research has already demonstrated some interesting applications, and in the last 5 years nanomaterials have begun entering clinical trials (eg, gold nanoshells, gold nanoparticles). The major limitation of this field is the unknown toxicology of nanomaterials, but this is a growing sub-discipline of nanotechnology and many questions will be answered in the next few years.

Figure 1.

An example of the different surface chemistry design of nanoparticles for biomedical applications. The base particles are usually designed with organic functional groups (eg, amines and carboxylic acids) that allow the particles to be conjugated to targeting agents (eg, antibodies, aptamers), polymers (eg, polyethylene glycol [PEG]), cell-penetrating peptides (eg, TAT peptides), or imaging agents (eg, fluorophores, radiolabels). In other nanoparticle designs, the nanoparticles has a porous interior that can store therapeutic agents.

Figure 1.

An example of the different surface chemistry design of nanoparticles for biomedical applications. The base particles are usually designed with organic functional groups (eg, amines and carboxylic acids) that allow the particles to be conjugated to targeting agents (eg, antibodies, aptamers), polymers (eg, polyethylene glycol [PEG]), cell-penetrating peptides (eg, TAT peptides), or imaging agents (eg, fluorophores, radiolabels). In other nanoparticle designs, the nanoparticles has a porous interior that can store therapeutic agents.

Close modal
Figure 2.

Size dependent optical effects of semiconductor nanoparticles. Semiconductor nanoparticles contain size dependent electronic and optical properties. A series of five different sized ZnS-capped CdSe semiconductor nanoparticles called quantum dots is used to demonstrate this principle. When these quantum dots are 2 nm, they emit in the blue but at 6 nm, they emit in the red. The size is inversely related the bandgap energy of the nanoparticle. This bandgap energy dictates the fluorescence emission.

Figure 2.

Size dependent optical effects of semiconductor nanoparticles. Semiconductor nanoparticles contain size dependent electronic and optical properties. A series of five different sized ZnS-capped CdSe semiconductor nanoparticles called quantum dots is used to demonstrate this principle. When these quantum dots are 2 nm, they emit in the blue but at 6 nm, they emit in the red. The size is inversely related the bandgap energy of the nanoparticle. This bandgap energy dictates the fluorescence emission.

Close modal
Figure 3.

Size-dependent plasmonic properties of metal nanoparticles. Extinction spectra and representative TEM images (inset) for a) 15, b) 30, c) 60, and d) 100 nm colloidal gold in solution. The plasmon resonance peak red-shifts and broadens at larger diameters, while the particle morphology remains quasi-spherical. Of note, the AU refers to arbitrary units.

Figure 3.

Size-dependent plasmonic properties of metal nanoparticles. Extinction spectra and representative TEM images (inset) for a) 15, b) 30, c) 60, and d) 100 nm colloidal gold in solution. The plasmon resonance peak red-shifts and broadens at larger diameters, while the particle morphology remains quasi-spherical. Of note, the AU refers to arbitrary units.

Close modal

Disclosures
 Conflict-of-interest: The authors declare no competing financial interests.
 Off-label drug use: None disclosed.

1
Smith AM, Mohs AM, Nie S. Tuning the optical and electronic properties of colloidal nanocrystals by lattice strain.
Nat Nanotechnol
.
2009
;
4
:
56
–63.
2
Peng X, Manna L, Yang W, et al. Shape control of CdSe nanocrystals.
Nature
.
2000
;
404
:
59
–61.
3
Li JJ, Wang YA, Guo W, et al. Large-scale synthesis of nearly monodisperse CdSe/CdS core/shell nanocrystals using air-stable reagents via successive ion layer adsorption and reaction.
J Am Chem Soc
.
2003
;
125
:
12567
–12575.
4
Murray CB, Kagan CR, Bawendi MG. Synthesis and characterization of monodisperse nanocrystals and close-packed nanocrystal assemblies.
Ann Rev Materials Sci
.
2000
;
30
:
545
–610.
5
Klostranec JM, Xiang Q, Farcas GA, et al., Convergence of quantum dot barcodes with microfluidics and signal processing for multiplexed high-throughput infectious disease diagnostics.
Nano Lett
.
2007
;
7
:
2812
–2818.
6
Yezhelyev MV, Al-Hajj A, Morris C, et al. In situ molecular profiling of breast cancer biomarkers with multicolor quantum dots. Advanced Materials.
2007
.19:3146–+.
7
Weissleder R, Kelly K, Sun EY, Shtatland T, Josephson L. Cell-specific targeting of nanoparticles by multivalent attachment of small molecules.
Nat Biotechnol
.
2005
;
23
:
1418
–1423.
8
Selvan ST, Patra PK, Ang CY, Ying JY. Synthesis of silica-coated semiconductor and magnetic quantum dots and their use in the imaging of live cells.
Angew Chem Int Ed Engl
.
2007
;
46
:
2448
–2452.
9
Gao XH, Cui Y, Levenson R, Chung L, Nie S. In vivo cancer targeting and imaging with semiconductor quantum dots.
Nat Biotechnol
.
2004
;
22
:
969
–976.
10
Tan WB, Jiang S, Zhang Y. Quantum-dot based nanoparticles for targeted silencing of HER2/neu gene via RNA interference.
Biomaterials
.
2007
;
28
:
1565
–1571.
11
Jiang W, Kim BY, Rutka JT, Chan WC. Nanoparticle-mediated cellular response is size-dependent.
Nature Nanotechnology
.
2008
;
3
:
145
–150.
12
Bagalkot V, Zhang L, Levy-Nissenbaum E, et al. Quantum dot-aptamer conjugates for synchronous cancer imaging, therapy, and sensing of drug delivery based on bi-fluorescence resonance energy transfer.
Nano Lett
.
2007
;
7
:
3065
–3070.
13
Chen J, Saeki F, Wiley BJ, et al. Gold nanocages: bioconjugation and their potential use as optical imaging contrast agents.
Nano Lett
.
2005
;
5
:
473
–477.
14
Gobin AM, Lee MH, Halas NJ, James WD, Drezek RA, West JL. Near-infrared resonant nanoshells for combined optical imaging and photothermal cancer therapy.
Nano Lett
.
2007
;
7
:
1929
–1934.
15
Fu A, Gu W, Boussert B, et al. Semiconductor quantum rods as single molecule fluorescent biological labels.
Nano Lett
.
2007
;
7
:
179
–182.
16
Xing Y, Chaudry Q, Shen C, et al. Bioconjugated quantum dots for multiplexed and quantitative immunohistochemistry.
Nat Protoc
.
2007
;
2
:
1152
–1165.
17
Goldman ER, Anderson GP, Tran PT, Mattoussi H, Charles PT, Mauro JM. Conjugation of luminescent quantum dots with antibodies using an engineered adaptor protein to provide new reagents for fluoroimmunoassays.
Anal Chem
.
2002
;
74
:
841
–847.
18
Gupta M, Caniard A, Touceda-Varek A, Campopiano DJ, Mareque-Rivas JC. Nitrilotriacetic acid-derivatized quantum dots for simple purification and site-selective fluorescent labeling of active proteins in a single step.
Bioconj Chem
.
2008
;
19
:
1964
–1967.
19
Howarth M, Takeo K, Kayashi Y, Ting AY. Targeting quantum dots to surface proteins in living cells with biotin ligase.
Proc Natl Acad Sci U S A
.
2005
;
102
:
7583
–7588.
20
Anderson RE, Chan WC. Systematic investigation of preparing biocompatible, single, and small ZnS-Capped CdSe quantum dots with amphiphilic polymers.
ACS Nano
.
2008
;
2
:
1341
–1352.
21
Mancini MC, Kairdolf BA, Smith AM, Nie S. Oxidative quenching and degradation of polymer-encapsulated quantum dots: new insights into the long-term fate and toxicity of nanocrystals in vivo.
J Am Chem Soc
.
2008
;
130
:
10836
–10837.
22
Dwarakanath S, Bruno JG, Shastry A, et al. Quantum dot-antibody and aptamer conjugates shift fluorescence upon binding bacteria.
Biochem Biophys Res Commun
.
2004
;
325
:
739
–743.
23
Fernández-Argüelles MT, Costa-Fernández JM, Pereiro R, Sanz-Medel A. Simple bio-conjugation of polymer-coated quantum dots with antibodies for fluorescence-based immunoassays.
Analyst
.
2008
;
133
:
444
–447.
24
Smith AM, Duan H, Mohs AM, Nie S. Bioconjugated quantum dots for in vivo molecular and cellular imaging.
Adv Drug Deliv Rev
.
2008
;
60
:
1226
–1240.
25
Nie S, Xing Y, Kim GJ, Simons JW. Nanotechnology applications in cancer.
Annu Rev Biomed Eng
.
2007
;
9
:
257
–88.
26
Smith AM, Dave S, Nie S, True L, Gao X. Multicolor quantum dots for molecular diagnostics of cancer.
Expert Rev Mol Diagn
.
2006
;
6
:
231
–244.
27
Chan WC, Maxwell DJ, Gao X, Bailey RE, Han M, Ne S. Luminescent quantum dots for multiplexed biological detection and imaging.
Curr Opin Biotechnol
.
2002
;
13
:
40
–46.
28
Klostranec JM, Chan WC. Quantum dots in biological and biomedical research: Recent progress and present challenges.
Advanced Materials
.
2006
;
18
:
1953
–1964.
29
Alivisatos AP. Semiconductor clusters, nanocrystals, and quantum dots.
Science
.
1996
;
271
:
933
–937.
30
Burda C, Chen X, Narayanan R, El-Sayed MA. Chemistry and properties of nanocrystals of different shapes.
Chem Rev
.
2005
;
105
:
1025
–1102.
31
Heath JR. The chemistry of size and order on the nanometer-scale.
Science
.
1995
;
270
:
1315
–1316.
32
Michalet X, Pinaud FF, Bentolila LA, et al. Quantum dots for live cells, in vivo imaging, and diagnostics.
Science
,
2005
;
307
:
538
–544.
33
Resch-Genger U, Grabolle M, Cavaliere-Jaricot S, Nitschke R, Nann T. Quantum dots versus organic dyes as fluorescent labels.
Nat Methods
.
2008
;
5
:
763
–775.
34
Alivisatos AP, Gu WW, Larabell C. Quantum dots as cellular probes.
Ann Rev Biomed Engineer
.
2005
;
7
:
55
–76.
35
Frangioni JV. In vivo near-infrared fluorescence imaging.
Curr Opin Chem Biol
.
2003
;
7
:
626
–634.
36
Wu XY, Liu H, Liu J, et al. Immunofluorescent labeling of cancer marker Her2 and other cellular targets with semiconductor quantum dots (vol 21, pg 41, 2003).
Nat Biotechnol
.
2003
;
21
:
452
–452.
37
Goldman ER, Clapp AR, Anderson GP, et al. Multiplexed toxin analysis using four colors of quantum dot fluororeagents.
Anal Chem
.
2004
;
76
:
684
–688.
38
Goldman ER, Medintz IL, Mattoussi H. Luminescent quantum dots in immunoassays.
Anal Bioanal Chem
.
2006
;
384
:
560
–563.
39
Ness JM, Akhtar RS, Latham CB, Roth KA. Combined tyramide signal amplification and quantum dots for sensitive and photostable immunofluorescence detection.
J Histochem Cytochem
.
2003
;
51
:
981
–987.
40
Pinaud F, King D, Moore HP, Weiss S. Bioactivation and cell targeting of semiconductor CdSe/ZnS nanocrystals with phytochelatin-related peptides.
J Am Chem Soc
.
2004
;
126
:
6115
–6123.
41
Rosenthal SJ, Tomlinson I, Adkins EM, et al. Targeting cell surface receptors with ligand-conjugated nanocrystals.
J Am Chem Soc
.
2002
;
124
:
4586
–4594.
42
Ghazani AA, Lee JA, Klostranec J,et al. High throughput quantification of protein expression of cancer antigens in tissue microarray using quantum dot nanocrystals.
Nano Lett
.
2006
;
6
:
2881
–2886.
43
Jaiswal JK, Goldman ER, Mattoussi H, Simon SM. Use of quantum dots for live cell imaging.
Nat Methods
.
2004
;
1
:
73
–78.
44
Lidke DS, Nagy P, Barisas BG, et al. Quantum dot ligands provide new insights into erbB/HER receptor-mediated signal transduction.
Nat Biotechnol
.
2004
;
22
:
198
–203.
45
Dahan M, Lévi S, Luccardini C, Rostaing P, Riveau B, Triller A. Diffusion dynamics of glycine receptors revealed by single-quantum dot tracking.
Science
.
2003
;
302
:
442
–445.
46
Le Gac S, Vermes I, van den Berg A. Quantum dots based probes conjugated to annexin V for photostable apoptosis detection and imaging.
Nano Lett
.
2006
;
6
:
1863
–1869.
47
Rosen AB, Kelly DJ, Schuldt AJ, et al. Finding fluorescent needles in the cardiac haystack: tracking human mesenchymal stem cells labeled with quantum dots for quantitative in vivo three-dimensional fluorescence analysis.
Stem Cells
.
2007
;
25
:
2128
–2138.
48
Dubertret B, Skourides P, Norris DJ, Noireaux V, Brivanlou AH, Libchaber A. In vivo imaging of quantum dots encapsulated in phospholipid micelles.
Science
.
2002
;
298
:
1759
–1762.
49
Ballou B, Ernst LA, Andreko S, et al. Sentinel lymph node imaging using quantum dots in mouse tumor models.
Bioconjug Chem
.
2007
;
18
:
389
–396.
50
Voura EB, Jaiswal JK, Mattoussi H, Simon SM. Tracking metastatic tumor cell extravasation with quantum dot nanocrystals and fluorescence emission-scanning microscopy.
Nat Med
.
2004
;
10
:
993
–998.
51
El-Sayed MA. Some interesting properties of metals confined in time and nanometer space of different shapes.
Acc Chem Res
.
2001
;
34
:
257
–264.
52
Link S, Mohamed MB, El-Sayed MA Simulation of the optical absorption spectra of gold nanorods as a function of their aspect ratio and the effect of the medium dielectric constant.
J Phys Chem B
.
1999
;
103
:
3073
–3077.
53
Link S, El-Sayed MA. Size and temperature dependence of the plasmon absorption of colloidal gold nanoparticles.
J Phys Chem B
.
1999
;
103
:
4212
–4217.
54
Jain PK, El-Sayed IH, El-Sayed MA. Au nanoparticles target cancer.
Nano Today
.
2007
;
2
:
18
–29.
55
Jain PK, Huang X, El-Sayed IH, El-Sayed MA. Noble metals on the nanoscale: optical and photothermal properties and some applications in imaging, sensing, biology, and medicine.
Acc Chem Res
.
2008
;
41
:
1578
–1586.
56
Englebienne P. Use of colloidal gold surface plasmon resonance peak shift to infer affinity constants from the interactions between protein antigens and antibodies specific for single or multiple epitopes.
Analyst
.
1998
;
123
:
1599
–1603.
57
Lyon LA, Musick MD, Natan MJ. Colloidal Au-enhanced surface plasmon resonance immunosensing.
Anal Chem
.
1998
;
70
:
5177
–5183.
58
Mayer KM, Lee S, Liao H, et al. A label-free immunoassay based upon localized surface plasmon resonance of gold nanorods.
ACS Nano
.
2008
;
2
:
687
–692.
59
Elghanian R, Storhoff JJ, Mucic RC, Letsinger RL, Mirkin CA. Selective colorimetric detection of polynucleotides based on the distance-dependent optical properties of gold nanoparticles.
Science
.
1997
;
277
:
1078
–1081.
60
Nam JM, Thaxton CS, Mirkin CA. Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins.
Science
.
2003
;
301
:
1884
–1886.
61
Rosi NL, Giljohann DA, Thaxton CS, Lytton-Jean AK, Han MS, Mirkin CA. Oligonucleotide-modified gold nanoparticles for intracellular gene regulation.
Science
.
2006
;
312
:
1027
–1030.
62
Park SJ, Taton TA, Mirkin CA. Array-based electrical detection of DNA with nanoparticle probes.
Science
.
2002
;
295
:
1503
–1506.
63
Han MS, Lutton-Jean AK, Oh BK, Heo J, Mirkin CA. Colorimetric screening of DNA-binding molecules with gold nanoparticle probes.
Angew Chem-Int Ed Engl
.
2006
;
45
:
1807
–1810.
64
El-Sayed IH, Huang XH, 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.
65
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
.
2003
;
63
:
1999
–2004.
66
Hauck TS, Jennings TL, Yatsenko T, Kumarades C, Chan WCW. Enhancing the toxicity of cancer chemo-therapeutics with gold nanorod hyperthermia.
Adv Mater
.
2008
;
20
:
3832
.
67
Chithrani BD, Chan WCW. Elucidating the mechanism of cellular uptake and removal of protein-coated gold nanoparticles of different sizes and shapes.
Nano Lett
.
2007
;
7
:
1542
–1550.
68
Chithrani BD, Ghazani AA, Chan WCW, Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells.
Nano Lett
.
2006
;
6
:
662
–668.
69
Pan Y, Neuss S, Leifert A, et al. Size-dependent cytotoxicity of gold nanoparticles.
Small
.
2007
;
3
:
1941
–1949.
70
Cedervall T, Lynch I, Foy M, et al. Detailed identification of plasma proteins adsorbed on copolymer nanoparticles.
Angew Chem-Int Ed Engl
.
2007
;
46
:
5754
–5756.
71
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
:
2050
–2055.
72
Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts.
Proc Natl Acad Sci U S A
.
2008
;
105
:
14265
–14270.
73
Derfus AM, Chan WCW, Bhatia SN. Probing the cytotoxicity of semiconductor quantum dots.
Nano Lett
.
2004
;
4
:
11
–18.
74
Magrez A, Kasas S, Salicio V, et al. Cellular toxicity of carbon-based nanomaterials.
Nano Lett
.
2006
;
6
:
1121
–1125.
75
Pisanic TR 2nd, Blackwell JD, Shubayer VI, Fiñones RR, Jin S. Nanotoxicity of iron oxide nanoparticle internalization in growing neurons.
Biomaterials
.
2007
;
28
:
2572
–2581.
76
Poland CA, Duffin R, Kinloch I, et al. Carbon nanotubes introduced into the abdominal cavity of mice show asbestos-like pathogenicity in a pilot study.
Nat Nanotechnol
.
2008
;
3
:
423
–428.
77
Liu Z, Davis C, Cai W, He L, Chen X, Dai H. Circulation and long-term fate of functionalized, biocompatible single-walled carbon nanotubes in mice probed by Raman spectroscopy.
Proc Natl Acad Sci U S A
.
2008
;
105
:
1410
–1415.
78
Schipper ML, Nakayama-Ratchford N, Davis CR, et al. A pilot toxicology study of single-walled carbon nanotubes in a small sample of mice.
Nat Nanotechnol
.
2008
;
3
:
216
–21.
79
Choi HS, Liu W, Misra P, et al. Renal clearance of quantum dots.
Nat Biotechnol
.
2007
;
25
:
1165
–1170.
80
Hardman R. A toxicologic review of quantum dots: toxicity depends on physicochemical and environmental factors.
Environ Health Perspect
.
2006
;
114
:
165
–172.

Author notes

1

Institute of Biomaterials & Biomedical Engineering, Donnelly Centre for Cellular and Biomolecular Research, Department Materials Science and Engineering, Department of Chemical Engineering, University of Toronto, Toronto, Ontario, Canada