455
Views
232
CrossRef citations to date
0
Altmetric
Review

Gold nanoparticles: From nanomedicine to nanosensing

, &
Pages 45-65 | Published online: 02 Nov 2008

Abstract

Because of their photo-optical distinctiveness and biocompatibility, gold nanoparticles (AuNPs) have proven to be powerful tools in various nanomedicinal and nanomedical applications. In this review article, we discuss recent advances in the application of AuNPs in diagnostic imaging, biosensing and binary cancer therapeutic techniques. We also provide an eclectic collection of AuNPs delivery strategies, including assorted classes of delivery vehicles, which are showing great promise in specific targeting of AuNPs to diseased tissues. However, successful clinical implementations of the promised applications of AuNPs are still hampered by many barriers. In particular, more still needs to be done regarding our understanding of the pharmacokinetics and toxicological profiles of AuNPs and AuNPs-conjugates.

Introduction

Some of the challenges facing conventional therapies are poor bioavailability and intrinsic toxicity. These have seriously compromised the therapeutic efficacy of many otherwise beneficial drugs. Nanoscopic systems that alter the pharmacological and therapeutic properties of molecules are being designed to overcome some of these limitations. Research efforts in this area have resulted in innovative nanodevices and nanostructures for use in applications such as diagnostics, biosensing, therapeutics, and drug delivery and targeting (CitationSahoo and Labhasetwar 2003; CitationFreitas 2005; CitationKawasaki and Player 2005; CitationKoo et al 2005; CitationCheng et al 2006; CitationPeters 2006; CitationBaron et al 2007; CitationVillalonga et al 2007; CitationHeath and Davis 2008).

Drug delivery with nanotechnological products takes advantage of pathophysiological conditions and anatomical changes within diseased tissues, compared with normal tissues, to achieve site-specific and targeted delivery (CitationSahoo et al 2007; CitationPrato et al 2008). Nanosystems are often accumulated at higher concentrations than normal drugs, thereby enhancing bioavailability at the targeted site. The enhanced drug targeting to the diseased tissues usually leads to reduced systemic toxicity. Moreover, incorporation of drug molecules in nanosized systems could improve drug solubility; offer a regulated drug release with enhanced retention at the target sites. These unique properties of nanosystems have been exploited to deliver drugs to harder-to-target sites such as the brain which offers a challenge due to the presence of the blood–brain barrier (CitationMaeda et al 2000; CitationMu and Feng 2003; CitationSahoo and Labhasetwar 2003; CitationTorchilin et al 2003; CitationRawat et al 2006; CitationSahoo et al 2007). Several varieties of engineered nanoparticles () have been widely used for drug delivery, imaging, biomedical diagnostics, and therapeutic applications (CitationSahoo and Labhasetwar 2003; CitationChen et al 2005; CitationLoo et al 2005; CitationYeh et al 2005; CitationHuang et al 2006; CitationLee and Wang 2006; CitationRawat et al 2006; CitationDong and Roman 2007; CitationLu et al 2007; CitationMaysinger 2007; CitationOyelere et al 2007; CitationSahoo et al 2007; CitationSkrabalak et al 2007; CitationCho et al 2008). Due to their small size (10 nm to 100 nm) (CitationSahoo et al 2007), several of these nanoparticles can penetrate smaller capillaries and are up taken by the cells. Many are also known to be biocompatible, undetected by the immune system, and biodegradable. Additionally, many could possess unique optical and electrical properties (CitationMaysinger 2007), key examples include quantum dots (Q-dots) and gold nanoparticles (AuNPs), making it possible to track their intracellular trafficking and localization (CitationMaysinger 2007; CitationOyelere et al 2007).

Table 1 Examples of nanoscale scaffolds for medical applications

For drug delivery applications, the drug of interest is either encapsulated, entrapped, adsorbed, attached, or dissolved into the nanoparticle matrix for release at the specific site (CitationSahoo and Labhasetwar 2003). An emerging trend in this field is the development of multifunctional nanoparticles. For example, polymeric micelles, which act as cancer-targets, drug delivery agents and possess magnetic resonance imaging (MRI) contrast characteristics, have been reported (CitationNasongkla et al 2006). CitationMajoros and colleagues (2006) have synthesized and characterized a multifunctional dendrimer conjugated with fluorescein isothiocyanate (for imaging), folic acid (for targeting cancer cells overexpressing folate receptors), and paclitaxel (chemotherapeutic drug).

Recent advances in the use of nanoparticles in medicine include delivery of antigens for vaccination (CitationPulliam et al 2007), gene delivery for treatment or prevention of genetic disorders (CitationRagusa et al 2007), and other therapeutics such as in cardiac therapy (CitationLanza et al 2006; CitationBrito and Amiji 2007), dental care (CitationBakó et al 2007), and orthopedic applications (CitationStreicher et al 2007). This has been the subject of many influential reviews (CitationLanza et al 2006; CitationHan et al 2007; CitationMorrow et al 2007; CitationSahoo et al 2007; CitationJain 2008). The focal point of the current review is to discuss the current use of AuNPs in medicine specifically, including targeted drug delivery, biosensing and bioimaging, and photothermal therapy.

Historic perspective on the use of AuNPs in medicine

Chrysotherapy, the use of gold in medicine, has been practiced since antiquity. Ancient cultures such as those in Egypt, India, and China used gold to treat diseases such as smallpox, skin ulcers, syphilis, and measles (CitationHuaizhi and Yuantao 2001; CitationRichards et al 2002; CitationGielen and Tiekink 2005; CitationKumar 2007). Presently, gold is in use in medical devices including pacemakers and gold plated stents (CitationEdelman et al 2001; CitationSvedman et al 2005), for the management of heart disease; middle ear gold implants (CitationThelen et al 2006), and gold alloys in dental restoration (CitationDemann et al 2005; CitationSvedman et al 2006). In the past few decades, several organogold complexes have emerged with promising antitumor, antimicrobial, antimalarial, and anti-HIV activities (CitationShaw 1999; CitationGielen and Tiekink 2005; CitationSun et al 2007). In fact, organogold compounds are now widely used for the treatment of rheumatoid arthritis (CitationShaw 1999; CitationMoolhuizen et al 2004; CitationSun et al 2007). Organogold compounds relieve arthritis symptoms such as joint pain, stiffness, swelling, bone damage, and also reduce the chance of joint deformity and disability. However, many of these compounds have shown reversible dose-dependent toxicities. In particular, at high doses, arthritis patients undergoing chrysotherapy often experience two common side effects: proteinuria and skin reactions (CitationMoolhuizen et al 2004).

Synthesis of AuNPs and its alloys

Several methods have been described in the literature for the synthesis of AuNPs of various sizes and shapes. The most popular synthetic method is by chemical reduction of gold salts such as hydrogen tetrachloroaurate (HAuCl4) using citrate as the reducing agent (CitationFrens 1973). This method produces monodisperse spherical AuNPs in the 10–20 nm diameter range. However, production of larger AuNPs (40–120 nm) by this method proceeds in low yields, often resulting in polydisperse particles. CitationBrown and Natan (1998) have reported the synthesis of monodisperse AuNPs with diameters between 30 and 100 nm using a seeding approach. The method is based on the use of the surface of AuNPs as a catalyst for the reduction of Au3+ by hydroxylamine. Subsequently, Murphy and colleagues employed this seed-mediated growth approach to control the shape and size of the nanoparticles (CitationJana et al 2001a, Citation2001b). Borohydride-reduced AuNPs seeds (3–4 nm diameter) were mixed with gold salt growth solution, rod-shaped micellar template (cetyltrimethylammonium bromide; CTAB), reducing agent (ascorbic acid), and small amount of silver ions for shape induction to produce spheroid or rod-like gold nanoparticles (CitationJana et al 2001a, Citation2001b). They have also improved this methodology to produce monodisperse, multiple-shaped AuNPs in higher yields than previously reported (CitationBusbee et al 2003; CitationSau and Murphy 2004).

Other methods for the synthesis of AuNPs include physical reduction (CitationSun et al 2003) (hollow Au nanostructures in large-scale), photochemical reduction (CitationKundu et al 2007) (cubic AuNPs), biological reduction (CitationMitra and Das 2008) (molecular hydrogels of peptide amphiphiles for producing various shapes of AuNPs), and solvent evaporation techniques (CitationPyrpassopoulos et al 2007) (2D Au super lattices). Recently, a simple and potentially cost effective microwave irradiation approach for the synthesis of shape-controlled AuNPs was reported (CitationKundu et al 2008). In this approach, irradiation of Au salt, reduced in CTAB micellar media, in the presence of alkaline 2,7-dihydroxy naphthalene (2,7-DHN), generate exclusively spherical, polygonal, rods, and triangular AuNPs within 90 seconds.

Bimetallic AuNPs, such as Au–Ag, have also attracted attention due to their interesting catalytic, structural and electronic properties, and the sensitivity of their surface plasmon resonance (SPR) properties (CitationHuang et al 2004; CitationLee and El-Sayed 2006). Accordingly, the development of simple and robust methods for the synthesis of bimetallic nanoparticles is currently of great interest. Spherical Au/Ag alloy nanoparticles whose SPR band could easily be tuned by varying the molar fractions of gold could be obtained by reduction of Au and Ag salt with sodium citrate in refluxing aqueous solution (CitationSun and Xia 2003). A seed-mediated approach (CitationLu et al 2002; CitationSun and Xia 2003) to synthesize Au-Ag core-shell nanorods from silver ions, using gold nanorods as seeds, has also been reported. Other methods for the synthesis of bimetallic AuNPs include sputter deposition technique in ionic liquids (CitationOkazaki et al 2008), photochemical synthesis (CitationPal and Esumi 2007), and deposition of Au/Ag on silica (CitationPal and De 2007). Relatively recently, a reverse microemulsion method to prepare silica-coated Au–Ag nanoparticles has been developed (CitationHan et al 2008).

Over the years, the ease of fabrication and the unique chemical and optical properties have sustained interests in the use of AuNPs in various molecular imaging and delivery applications. More significantly, the unique biodistribution of AuNPs within tumors have led to the discovery of gold-based nanosystems as delivery vehicles for chemotherapeutic agents (CitationPaciotti et al 2006).

Bioimaging

Researchers have used various exogeneous agents to visualize key subcellular compartments. Cell imaging is achieved through the generation of colorimetric contrast between different cells/subcellular organelles by these imaging agents. Conventional exogeneous imaging agents include lanthanide chelates and organic fluorophores (CitationSharma et al 2006). However, organic fluorophores are prone to photobleaching, low quantum yields, and broad emission window (CitationBruchez et al 1998; CitationChan et al 2002). Lanthanide chelates, on the other hand, are prone to nonselective localization in extravascular space (CitationSharma et al 2006). The shortcomings of the conventional imaging agents have limited their applications as biomedical diagnostic tools and have stimulated interest in typical nanomaterials, such as magnetic nanoparticles (CitationKim et al 2003; CitationMartina et al 2005; CitationLee et al 2006b), Q-dots (CitationAkerman et al 2002; CitationKim et al 2004; CitationGao et al 2005), and AuNPs (CitationBoyer et al 2002; CitationCognet et al 2003; CitationLoo et al 2005; CitationHuang et al 2006; CitationIpe et al 2006; CitationLewis et al 2006; CitationQian et al 2008) as alternative contrasting agents. These nanomaterials are optimal diagnostic tools since they eliminate most of the vulnerabilities of the conventional imaging agents. However, the intrinsic cytotoxicity of most nanomaterials has diminished their utility in many in vitro and in vivo application (CitationEl-Sayed et al 2005b; CitationThurn et al 2007; CitationLewinski et al 2008). AuNPs are unique exceptions because they are more tolerable and compatible with cellular environment (CitationTkachenko et al 2003; CitationConnor et al 2005; CitationShukla et al 2005; CitationPan et al 2007). In addition, the colorimetric contrast observed within the AuNPs treated cells could be controlled by size (CitationTurkevich et al 1951; CitationKreibig and Genzel 1985; CitationKhlebtsov et al 2005), shape (CitationSarkar and Halas 1997; CitationJin et al 2001; CitationMurphy and Jana 2002), or even surface modification (CitationMarinakos et al 1999; CitationCaruso and Antonietti 2001) of the AuNPs due to a phenomenon called surface plasmon resonance (SPR) (CitationSharma et al 2006). When excited, the SPR of AuNPs could scatter and/or absorb light in the visible or the near-infrared (NIR) spectrum (CitationJain et al 2006), an extremely useful property for in vivo optical imaging techniques such as photoacoustic (CitationAgarwal et al 2007), and two-photon luminescence imaging (CitationDurr et al 2007). These two optical diagnostic techniques specifically generate cellular contrast by tuning the SPR of the AuNPs to the NIR spectrum. Other noninvasive diagnostic tools such as MRI (CitationDebouttiere et al 2006) and X-ray computed tomography (X-ray CT) (CitationKim et al 2007) have utilized AuNPs as contrasting agent due to the ease of surface modification and higher X-ray absorption coefficient, respectively.

Magnetic resonance imaging

Magnetic resonance imaging is a noninvasive diagnostic tool that applies magnetic fields to the heterogeneous composition of water in organisms (CitationWeissleder and Mahmood 2001; CitationCaravan et al 2003; CitationLangereis et al 2004). Different water proton relaxivity rates translate into contrasting images of different cells (CitationDebouttiere et al 2006). The MRI images can be enhanced by reducing the longitudinal and transverse relaxation time of the water proton (CitationCaravan et al 1999; CitationMerbach and Toth 2001). The enhancement is often observed by the use of contrasting agents such as gadolinium chelates (CitationCaravan et al 1999) or superparamagnetic iron oxide (CitationAime et al 1998). The most widely used contrasting agent for MRI is gadolinium-diethyltriaminepentaacetic acid (Gd-DTPA) (CitationSharma et al 2006). In this reagent, GdIII is the contrasting agent, while DTPA is the chelating ligand that forms a complex with GdIII to minimize the leaching of the cytotoxic, ionic GdIII into the cellular milieu (). Despite the contrast enhancement, the imaging application of Gd-DTPA is still hampered by their rapid renal clearance (CitationDebouttiere et al 2006). For optimal contrast enhancement, AuNPs have been utilized as a delivery vehicle to convey multiple Gd-DTPA complexes into selective cellular targets. Dithiolated DTPA (DTDTPA) has been utilized in place of DTPA to chelate to ionic GdIII and permit conjugation onto 2 to 2.5 nm AuNPs surface (). In the MRI study performed by Roux and colleagues, Gd-DTDTPA/AuNPs conjugates retain the intrinsic contrasting property of Gd-DTPA under MRI and provide the desired contrast enhancement compared to single Gd-DTPA (CitationDebouttiere et al 2006). However, the in vivo application and cytotoxicity of the Gd-DTDTPA/AuNPs conjugates have not been fully investigated (CitationDebouttiere et al 2006).

Figure 1 Chemical structures of DTPA (left), and DTDTPA (right).

Figure 1 Chemical structures of DTPA (left), and DTDTPA (right).

Figure 2 Schematic illustration of DTDTPA–AuNPs.

Figure 2 Schematic illustration of DTDTPA–AuNPs.

X-ray computed tomography

X-ray computed tomography is another noninvasive diagnostic method that generates three-dimensional images of different cells based on a series of two-dimensional X-ray images compiled around a single rotating axis (CitationNCI 2003). Contrasting agents are often utilized to enhance the contrast between cells because of their affinity to absorb X-rays. One of the widely used contrasting agents in X-ray CT is called Ultravist (iopromide), an iodinated small molecule dye () (CitationKim et al 2003). There are several shortcomings of Ultravist that includes renal toxicity (CitationHizoh and Haller 2002; CitationHaller and Hizoh 2004), vascular permeation, and limited imaging interval due to rapid renal execretion (CitationKim et al 2007). The limitations observed in current CT contrasting agents were recently overcome by the use of AuNPs. AuNPs present several advantages over the current contrasting agents, such as higher X-ray absorption coefficients (CitationHainfeld et al 2006), versatility in surface modification, and regulated control in the size and shape of the AuNPs. Recently, CitationKim and colleagues (2007) performed CT studies on AuNPs coated with poly-ethylene gycol (PEG) (CitationAllen et al 1991; CitationPapahadjopoulos et al 1991; CitationHerrwerth et al 2003; CitationZheng et al 2003; CitationBallou et al 2004; CitationKohler et al 2004; CitationLee et al 2006a) as antibiofouling agents, to test their in vivo application as CT contrast agents for angiography and hepatoma detection. X-ray absorption coefficient measurements in vitro revealed that the attenuation of PEG-coated AuNPs is 5.7 times higher than Ultravist at equal concentration. The PEG-coated AuNPs have a longer blood circulation time, approximately 4 hours without apparent loss of contrast in a mice model, compared with only about 10 minutes for Ultravist. Also, a two-fold contrast enhancement was seen between the hepatoma and its surrounding healthy liver cells for up to 24 hours. These results showed the feasibility of AuNPs as a CT contrast agent in vivo. Although no considerable toxicity was detected in liver cells (HepG2) upon exposure to the PEG-coated AuNPs for 24 hours, further studies need to be undertaken for PEG-coated AuNPs to be considered a clinically useful contrast agent.

Figure 3 Chemical structure of Ultravist (iopromide), a contrast agent used in X-ray computed tomography.

Figure 3 Chemical structure of Ultravist (iopromide), a contrast agent used in X-ray computed tomography.

Optical imaging

In photoacoustics (CitationAgarwal et al 2007) and two-photon luminescence (CitationDurr et al 2007), AuNPs are utilized as contrasting agent that permits light scattering and/or absorption at the NIR spectrum (between 700–1000 nm) (CitationAgarwal et al 2007). This imaging window is known as the “tissue transparency window”. Light penetration at this imaging window is at maximum with minimum loss to hemoglobin and water absorption, thereby permitting deep imaging of the cells (CitationMahmood and Weissleder 2003). CitationAgarwal and colleagues (2007) used 15-nm AuNPs in a photoacoustic experiment to enhance cell contrast upon irradiation by a short pulse laser. The acoustic emissions created by the AuNPs are collected by ultrasonic array to recreate the initial heat distribution that images the target cell. AuNPs efficiently emit two-photon luminescence because they can sustain SPR with little or no damping after the photon excitation (CitationSonnichsen et al 2002). The two-photon cross sections of AuNPs have been exploited in two-photon luminescence experiments to image target cells (CitationWang et al 2005). With appropriate delivery platforms on the AuNPs, photoacoustics (CitationAgarwal et al 2007) and two-photo luminescence imaging (CitationDurr et al 2007) have been used to selectively image LnCAP prostate cancer and A431 skin cancer cells, respectively. Most of the delivery platforms in optical diagnostic application are protein- and peptide-based, these will be discussed later in this review.

Biosensing

Biosensors employ biological molecules such as antibodies, enzymes, carbohydrates, and nucleic acids to identify or follow the course of any biological phenomena of interest (CitationOtsuka et al 2001; CitationMcFadden 2002). Interactions, such as hydrogen bonding and charge–charge transfers between the ligand and receptor molecules, coupled with read-out techniques such as colorimetry, fluorescence, biomagnetic signals, etc, are used for sensing specific biochemical events (CitationMcFadden 2002). Biosensors are finding use in various applications: food processing, to monitor food-borne pathogens in the food supply; environmental monitoring, to detect pollutants and pesticides in the environment; biowarfare defense, to detect bacteria, viruses and biological toxins; and clinical diagnostics, to measure blood glucose levels (CitationMcFadden 2002; CitationLi and Rothberg 2004).

AuNPs exhibit special optical and electronic properties such as enhanced SPR, surface-enhanced emission, and surface-enhanced Raman scattering (SERS) (CitationFrederix et al 2003; CitationHuang et al 2007a). These properties have been used in sensing and/or monitoring numerous molecular events including protein–protein interaction, protein aggregation, and protein folding (CitationDe et al 2007; CitationGhoshmoulick et al 2007; CitationVillalonga et al 2007). For example, the SPR signals of AuNPs have been used not only to selectively detect DNAs but also to differentiate between perfect and mismatched DNA duplex. Mirkin and colleagues reported that mercaptoalkyloligonucleotide-modified AuNPs probes generate cross-linked polymeric aggregates that signaled hybridization with complementary oligonucleotide target via color change (CitationElghanian et al 1997). The color of the nanoparticle aggregates appeared to vary as the interparticle distance changes; a phenomenon attributed to the SPR of Au (CitationElghanian et al 1997). Moreover, the colorimetric transition temperatures of the nanoparticle aggregates were used to differentiate a perfect match target from a mismatch base target. However, this approach is limited in that it is inherently a one-color system that is based on a gray scale. A system that overcomes this handicap, by performing multiplexed detection of oligonucleotide targets, has been reported (CitationCao et al 2002). This system consists of 13-nm AuNPs probes functionalized with oligonucleotides and Raman-dye labels as Raman spectroscopic fingerprint. It distinguishes between oligonucleotide sequences using Ag surface-enhancement of SERS as readout. Several dissimilar DNA targets and two RNA targets were distinguished (CitationCao et al 2002).

A new aggregation phenomenon of DNA-functionalized AuNPs, induced by noncross-linking target DNA hybridization, has been recently reported. This phenomenon allows simple and rapid colorimetric sensing of DNA hybridization that is sufficiently sensitive to detect terminal single-base-pair mismatches (CitationSato et al 2003, Citation2005; CitationLi and Rothberg 2004). Based on its simplicity and easy read-out (CitationSato et al 2003), this technique has opened up a new possibility for reliable genetic diagnosis. Another colorimetric hybridization assay that uses unmodified/unfunctionalized AuNPs for sequence-specific detection of DNA has appeared in the literature (CitationLi and Rothberg 2004). Based on the differences in electrostatic properties between single stranded DNA (ssDNA) and double stranded DNA (dsDNA), ssDNA selectively adsorbs on and stabilizes the AuNPs against aggregation in high salt buffers relative to dsDNA. This assay has an added advantage of being completely independent of the detection step while adaptable to sensing single-base-pair mismatches between probe and target.

AuNPs colorimetric response to changes in environment has been extended to detection of protein–ligand interactions (CitationTsai et al 2005). For example, concanavalin (ConA)–mannose interaction has been investigated using mannose modified–AuNPs (Man–AuNPs). It was demonstrated that the interaction between ConA and Man–AuNPs resulted in aggregation (blue colored aggregates) suggesting specific binding of Man–AuNPs to ConA. To further probe the specificity of this interaction, a variety of proteins were added to the Man–AuNPs/ConA aggregate, and it was shown, via colorimetric response (blue to burgundy = loss of aggregation), that a subset of these proteins effectively compete with ConA for Man–AuNPs binding. The system is sensitive within a nanomolar range and potentially could be applied to investigate a broad range of protein–ligand interactions.

AuNPs/enzymes-based biosensors that measure cellular glucose levels have also been developed for potential use in diabetes management (CitationStonehuerner et al 1992; CitationAubin et al 2005; CitationHa et al 2005; CitationSimonian et al 2005; CitationHill and Shear 2006; CitationJena and Raj 2006; CitationZhao et al 2008). These sensors use glucose oxidase immobilized on AuNPs to detect glucose concentrations (CitationPandey et al 2007). AuNPs immobilization of glucose oxidase resulted in glucose sensors with enhanced sensitivity and stability. Using AuNPs to which a yeast iso-1-cytochrome c (Cytc) is covalently attached, Zare and collegues have demonstrated that AuNPs could be used as a colorimetric sensor to follow the folding or unfolding of an appended protein molecule (CitationChah et al 2005). Upon exposure to buffers of different pH, the appended Cytc unfolds at low pH, thus inducing AuNPs aggregation while refolding at high pH, results in the loss of aggregation. These conformational changes caused measurable shifts in the AuNPs’ color and could be detected by UV–VIS absorption spectroscopy (CitationChah et al 2005). In a similar manner, the pH dependent shifts in the AuNPs plasmon resonance have recently been used to track protein structural changes induced by glycation (CitationGhoshmoulick et al 2007), a modification that is of importance in the clinicopathology of diabetes (CitationHudson et al 2002). Glycation progress was found to correlate with a significant shift in the size distribution of AuNPs as well as their plasmon resonance peak and intensity.

Photothermal therapy

Photothermal therapy is a less invasive experimental technique that holds great promise for the treatment of cancer and related disease conditions (CitationHuang et al 2006). It combines two key components: (i) light source, specifically lasers with a spectral range of 650–900 nm (CitationHuang et al 2006) for deep tissue penetration, and (ii) optical absorbing AuNPs which transform the optical irradiation into heat on a picosecond time scale, thereby inducing photothermal ablation (CitationChen et al 2007a; CitationHaba et al 2007). Recent developments have shown that the spectral signature of AuNPs could be tailored or tuned by altering their shape or size. El-Sayed and colleagues have demonstrated that gold nanorods have a longitudinal absorption band in the NIR on account of their SPR oscillations and are effective as photothermal agents (CitationHuang et al 2006). Other gold nanostructures such as gold nanoshells (CitationLoo et al 2005), gold nanocages (CitationChen et al 2007a), and gold nanospheres (CitationHuang et al 2008) () have also demonstrated effective photothermal destruction of cancer cells and tissue. However, efficient in vivo targeting of AuNPs to heterogeneous population of cancer cells and tissue still requires better selectivity and noncytotoxicity to surrounding normal cells.

Figure 4 TEM images of plasmonic gold nanostructures commonly used for PPTT. a) nanospheres, b) nanorods, c) nanoshells.

Figure 4 TEM images of plasmonic gold nanostructures commonly used for PPTT. a) nanospheres, b) nanorods, c) nanoshells.

Selectivity

Although nanoparticle-based therapeutics exploits the enhanced permeability and retention (EPR) effect for delivery into tumors, not all tumors are amenable to this effect, especially in regard to the delivery of the nanoparticles of relatively large size (CitationIshida et al 1999). In addition, selective photothermolysis is not obtained for small tumors or single metastatic cells because heat diffusion from hot particles increases the damaged tissue area with longer exposure times (CitationZharov et al 2005). Hence, other methods of selective nanoparticle delivery need to be developed in order to achieve effective photothermal therapy. Recent studies have shown that AuNPs conjugated to antibodies (CitationHuang et al 2006) and viral vectors (CitationEverts et al 2006) could be used for selective and efficient photothermal therapy. CitationHuang and colleagues (2006) have demonstrated that gold-nanorods conjugated to anti-epidermal growth factor receptor (anti-EGFR) antibodies selectively target cell lines that overexpress EGFR. Subsequent continuous laser exposure of nanoparticle-treated cells resulted in photothermal destruction of the EGFR positive cells at half the energy required to kill EGFR negative cells. Similarly, treatment of breast cancer cell line overexpressing HER2 with HER2-targeted gold nanoshells (CitationHirsch et al 2003; CitationLowery et al 2006; CitationBernardi et al 2008) and nanocages (CitationChen et al 2007a) followed by exposure to laser light in the NIR has been shown to selectively induce cell death to the HER2 positive cell in vitro.

The potential of photothermal therapy in disease intervention has recently been extended to include parasite infections. Using gold nanorods conjugated with antibody selective for Toxoplasma gondii, CitationPissuwan and colleagues (2007) reported that plasmonic heating with a 650-nm laser at power density of 51 W/cm2 resulted in more than 80% destruction of T. gondii tachyzoites. This is one of the early examples of photothermal intervention in parasitic diseases; more studies need to be done to ascertain its general utility.

Toxicity

Organogold antiarthritis compounds, such as auranofin and Tauredon (), have presented some dose-dependent adverse side effects. Nevertheless, AuNPs are generally considered to be benign. However, the size similarity of AuNPs to biological matters could provide “camouflage” to cellular barriers, leading to undesired cellular entry which might be detrimental to normal cellular function (CitationConnor et al 2005). The prospect that the inadvertent AuNPs’ cellular entry could result in toxic side effects have stimulated intense efforts aimed at providing better insight into their toxicity profile. CitationPan and colleagues (2007) recently conducted a systematic investigation of the size-dependent cytotoxicity of water soluble, triphenylphosphine-stabilized AuNPs against four cell lines: Hela cervix carcinoma epithelial cells, Sk–Mel–28 melanoma cells, L929 mouse fibroblast, and J774A1 mouse monocytic/macrophage cells. They found that AuNPs 1 to 2 nm in size displayed cell-type dependent cyotoxicity with high micromolar IC50s. In contrast, AuNPs 15 nm in size were nontoxic to cells at concentrations 60-fold higher than the IC50 of the smaller AuNPs. These results seemed to confirm size-dependent toxicity of AuNPs (CitationPaciotti et al 2004; CitationEl-Sayed et al 2005a, Citation2005b; CitationDebouttiere et al 2006; CitationHuang et al 2006, Citation2007b; CitationVisaria et al 2006; CitationKim et al 2007; CitationNicholas et al 2007), an inference that has hitherto been somewhat ambivalent.

Figure 5 Examples of chrysotherapeutics utilized in rheumatoid arthritis treatment. Chemical structures of auranofin (left) and Tauredon (right).

Figure 5 Examples of chrysotherapeutics utilized in rheumatoid arthritis treatment. Chemical structures of auranofin (left) and Tauredon (right).

Earlier investigation by Rotello and colleagues have shown that cationic side chains (CTAB) tend to impart moderate toxicity to AuNPs whereas anionic side chains (carboxylate-derived) are generally nontoxic () (CitationGoodman et al 2004). Further analyses revealed that the toxicity observed with the cationic AuNPs is due to cell lysis rather than receptor-mediated endocytosis (RME). A later study by CitationConnor and colleagues (2005) revealed that the CTAB-bound AuNPs by themselves do not present measurable toxicity to K-562 leukemia cell line. Instead, toxicity was due to the presence of unbound CTABs. This result suggested that the toxicity observed in Rotello’s experiment could be the consequence of the unbound CTAB derivative.

Figure 6 Cationic (CTAB-derived, top right) and anionic (carboxylate-derived, bottom right) side chain surface modification of AuNPs.

Figure 6 Cationic (CTAB-derived, top right) and anionic (carboxylate-derived, bottom right) side chain surface modification of AuNPs.

Overall, current literature evidence support the assertion that AuNPs and their conjugates are relatively less toxic to cells (CitationTkachenko et al 2003; CitationConnor et al 2005; CitationShukla et al 2005; CitationPan et al 2007; CitationLewinski et al 2008). Nevertheless, identification of proper delivery platforms will further enhance the prospects of AuNPs as tools for noninvasive disease diagnosis and treatment.

Delivery

Efficient delivery of AuNPs into a living system requires overcoming natural biological barriers such as the cell membrane and the reticuloendothelial system (RES). For specific tumor targeting, AuNPs face additional challenges from receptor specificity and intratumor barriers. Potential approach for optimizing AuNPs delivery is particle size reduction (“true nanometer scale”) or acquisition of surface modification. For example, large AuNPs are quickly opsonized by blood and eliminated by the RES in mammalian cells (CitationWoodle et al 1994; CitationRaynal et al 2004; CitationRoger and Basu 2005; CitationPaciotti et al 2006). To bypass RES, antibiofouling agents such as thiol-derivatized poly-ethylene glycol (PEG-SH) have been grafted onto AuNPs surface as secondary coating. It has been observed that this secondary coating could delay RES clearance to liver from 0.5 hours to 72 hours in a mice model, an approximately 150-fold improvement compared with the unmodified CTAB-capped AuNPs (CitationNiidome et al 2006). Several investigators have grafted different delivery platforms onto AuNPs surface to attempt cellular selectivity, internalization, and localization within heterogeneous population of cancer cells in solid tumors. These delivery platforms generally consist of macromolecules such as proteins and peptides or small molecules such as folic acid and paclitaxel. Several of these platforms have shown very promising results in delivering AuNPs into solid tumors (CitationEl-Sayed et al 2005a, Citation2005b; Citationde la Furente et al 2006; CitationHuang et al 2006; CitationPaciotti et al 2006; CitationVisaria et al 2006).

Protein delivery platforms

Proteins such as tumor necrosis factor-α (TNFα) and anti-EGFR antibodies have been successfully grafted onto AuNPs surface and utilized in conjunction with hyperthermia to selectively kill cancer cells. Another protein that shows selective cellular intake into cancer cells when conjugated to AuNPs is transferrin. However, its therapeutic application is yet to be fully investigated (CitationYang et al 2005). In several of these protein–AuNPs conjugates, the protein component selectively penetrates cancer cells through RME (CitationPaciotti et al 2006; CitationVisaria et al 2006). TNFα provides an illustrative example in this regard.

TNFα is a potent cytokine that induces systemic inflammation. Additionally, TNFα is known to be overexpressed in solid tumors (CitationPaciotti et al 2004) and mediates hemorrhagic necrosis in solid tumors (CitationNorth and Havell 1988; CitationKircheis et al 2002; CitationPaciotti et al 2006; CitationVisaria et al 2006). The later property suggests TNFα may find use in cancer therapy. However, TNFα has low therapeutic index due to nonselective acute toxicity that results from cell exposure (CitationBrouckaert et al 1986; CitationHieber and Heim 1994). Recent observations on selective uptake of AuNPs by tumors have enabled a re-evaluation of the potential application of TNFα in cancer therapy. TNFα grafted onto AuNPs surface has reduced systemic toxicity compared to the native unconjugated TNFα. More importantly, the TNFα-AuNPs conjugates are able to accumulate preferentially in the tumor vasculature. The selective uptake of the AuNPs into the tumor has been suggested to be due to the leaky vasculature of the tumor blood vessels, which allows AuNPs of sizes ranging between 20 to 100 nm to passively diffuse into the tumor interstitium (CitationPaciotti et al 2004). A current example of chemotherapy agents based on this technology is CYT-6091 or Aurimune™ () developed by CytImmune (Rockville, MD, USA) and is about to enter into phase II clinical trials for the treatment of melanoma, colorectal cancer, and urinary tract cancer. Aurimune™ is a multivalent drug consisting of 33-nm colloidal AuNPs, onto which is grafted TNFα for specific solid-tumor targeting and thiol-derivatized poly-ethylene glycol to bypass RES. Upon tumor selective uptake, the drug is internalized into tumor cells through TNFα-mediated RME (CitationPaciotti et al 2006; CitationVisaria et al 2006). To confirm that TNFα-binding contributes to the selectivity observed toward MC-38 tumor cells, TNFα-resistant B16/F10 melanoma cells were exposed to Aurimune™. Only temporary growth inhibition of these TNFα-resistant melanoma cells was observed (CitationVisaria et al 2006). In addition to directing AuNPs to TNFα-specific tumor cells, TNFα also acts as an anticancer agent that induces hemorrhagic necrosis in solid tumors (CitationNorth and Havell 1988; CitationKircheis et al 2002; CitationPaciotti et al 2006; CitationVisaria et al 2006).

Figure 7 Illustration of TNFα/PEG-conjugated AuNPs (Aurimune-T™).

Figure 7 Illustration of TNFα/PEG-conjugated AuNPs (Aurimune-T™).

The effect of combination of Aurimune™ with hyperthermia on cancer cell viability has also been investigated. SCK murine mammary carcinoma cells were treated with Aurimune™ and heated to 42.5 °C. In both in vivo and in vitro tumor cell survival studies, Aurimune™ at 250 μg/kg together with hyperthermia was shown to possess 2- to 3-fold higher anticancer activity compared to Aurimune™ alone (CitationVisaria et al 2006). Possibly, the Aurimune™ hyperthermia effect induces the macro- and microvasculature shutdown (CitationSrinivasan et al 1990; CitationUmeno et al 1994) of the tumor cells, and consequently cuts off the blood flow that transports nutrients and oxygen to the tumors. Additionally, increase in anaerobic glycolysis in transformed cells that leads to high acidity and acidic byproducts buildup (CitationRaghunuand et al 2003), could enhance the susceptibility of tumor cells to heat shock that resulted from hyperthermia and expedite the tumor apoptosis.

Epidermal growth factor receptor is another receptor that is overexpressed in several types of cancer including lung and pancreatic cancers (CitationArteaga 2001; CitationXiong and Abbruzzese 2002; CitationPaez et al 2004; CitationAhmed and Salgia 2006; CitationPrudkin and Wistuba 2006; CitationCohenuram and Saif 2007). Overexpression of EGFR has been demonstrated to culminate from mutations on EGFR gene that often proceed to uncontrolled cell division and the proliferation of cancer cells (CitationLynch et al 2004). Two therapeutic approaches that target EGFR-enriched cancer cells are monoclonal antibody-based therapy and tyrosine kinase inhibitors (CitationAhmed and Salgia 2006; CitationPrudkin and Wistuba 2006; CitationCohenuram and Saif 2007). Monoclonal antibodies such as anti-EGFR antibody have been investigated as a possible anticancer therapy for lung cancer. Moreover, anti-EGFR antibody has been grafted onto 35 nm AuNPs and employed as a cancer diagnostic tool (CitationEl-Sayed et al 2005b) and for photothermal therapy (Huang et al 2007c) in an oral cancer model. Anti-EGFR–AuNPs conjugates designed for HSC and HOC oral cancer diagnostics utilized the color-scattering property of the AuNPs. When illuminated with a white light at specific angles, AuNPs, depending on their size and shape, will scatter light of many colors (CitationYguerabide and Yguerabide 1998). In a study aimed at diagnosis, CitationEl-Sayed and colleagues (2005b) found that anti-EGFR-AuNPs conjugates bound readily in a homogenous manner to both HOC and HSC oral cancer cell lines overexpressing EFGR. The binding of the anti-EGFR-AuNPs conjugates enabled a clear visualization of these cells under a microscope. However, HaCaT, a noncancerous cell line in which EFGR expression is depressed, only showed a random AuNPs conjugate binding. The random distribution of the conjugate leads to poor visualization, and individual HaCaT cells were undistinguishable. Such binding preferences, together with the unique light scattering property, provide a useful diagnostic tool to distinguish between noncancerous and cancerous cells. This concept has been successfully employed by CitationQian and colleagues (2008) to image EGFR-enriched Tu696 human head-and-neck carcinoma cells in vivo and in vitro using an ScFv version of the anti-EGFR antibody. Additionally, anti-EGFR–AuNPs conjugates have been used in photothermal therapy to target and selectively destroy EGFR-enriched cancers (CitationEl-Sayed et al 2005a; CitationHuang et al 2006, Citation2007b).

Peptide delivery platforms

Most peptides used for AuNPs delivery target the cell nucleus (). The nucleus is an attractive target for photothermal therapy because it contains the cellular genetic machinery. These nuclear membrane-penetrating peptides facilitate the entry of AuNPs into the nuclei by first permitting entry into the cell via RME followed by nuclear localization through interaction with the nuclear pore complex (CitationTkachenko et al 2003). Most of the nuclear membrane-penetrating peptides are derived from virus sources. Common examples include the Simian virus nuclear localization peptides (NLS) (CitationKalderon et al 1984; CitationTkachenko et al 2003; CitationOyelere et al 2007), HIV 1 Tat-protein-derived peptides (Citationde la Furente and Berry, 2005), and peptides derived from adenovirus fiber protein. Other nonnuclear targeting peptides that have been used as delivery vehicle for AuNPs include various forms of the RGD peptides (CitationTkachenko et al 2004; Citationde la Furente et al 2006).

Table 2 Peptides utilized in AuNPs delivery

NLS peptides are peptides utilized by viruses to cross many cellular membranes especially the nuclear membrane. Tkachenko and colleagues have described a series of NLS peptides that are grafted onto 20-nm AuNPs (CitationHayat 1989; CitationTkachenko et al 2003, Citation2004; CitationLiu et al 2007). The ability of these NLS-AuNPs conjugates to selectively accumulate into the nucleus was investigated in intact Hela, 3T3/NIH, and HepG2 cells (CitationTkachenko et al 2004). NLS peptides derived from the SV40 large T antigen successfully facilitates the entrance of the conjugate into the nucleus of HepG2 when directly injected inside the cytoplasm (CitationFeldherr and Akin 1990; CitationTkachenko et al 2003). However, the conjugates were trapped inside the cytoplasm when included in the cell growth media. This may be the result of endosome capturing of the conjugates after entrance into the cell via RME. Hence, nuclear targeting was not observed for all three cell lines (CitationTkachenko et al 2003, Citation2004). However, we recently discovered that when directly grafted onto AuNPs via a thioalkyl linker (), NLS derived from the SV40 Large T antigen efficiently facilitated nuclear delivery of AuNPs to HSC oral cancer cells and noncancerous human HaCaT cells () (CitationOyelere et al 2007). Such discrepancy in nuclear translocation could be due to the difference in cell types or AuNPs fabrication technique.

Figure 8 Illustration of SV40 Large T NLS conjugated to AuNPs surface via thiolalkyl linker.

Figure 8 Illustration of SV40 Large T NLS conjugated to AuNPs surface via thiolalkyl linker.

Figure 9 Dark field light scattering images of CTAB-capped Au-nanorods and SV40 largeT NLS/Au-nanorod conjugates after 2 h incubation with cells. A) CTAB-capped Au-nanorods in HaCaT normal cells. B) CTAB-capped Au-nanorods in HSC cancer cells. C) SV40 large T NLS/Au-nanorod conjugates in HaCaT normal cells. D) SV40 large T NLS/Au-nanorod conjugates in HSC cancer cells. Scale bar: 10 μm.

Figure 9 Dark field light scattering images of CTAB-capped Au-nanorods and SV40 largeT NLS/Au-nanorod conjugates after 2 h incubation with cells. A) CTAB-capped Au-nanorods in HaCaT normal cells. B) CTAB-capped Au-nanorods in HSC cancer cells. C) SV40 large T NLS/Au-nanorod conjugates in HaCaT normal cells. D) SV40 large T NLS/Au-nanorod conjugates in HSC cancer cells. Scale bar: 10 μm.

Peptides derived from adenovirus have also been used to promote nuclear penetration (CitationTkachenko et al 2003, Citation2004). The full, single fiber protein sequence from adenovirus contains both RME and NLS domains (CitationTkachenko et al 2003). Its AuNPs conjugate has been shown to successfully avoid the endosome and penetrates the nucleus of the HepG2 cells (CitationTkachenko et al 2003, Citation2004). Moreover, individual RME and NLS domains derived from the adenovirus fiber protein have been independently investigated for nuclear penetration. Not surprisingly, adenoviral RME only permitted cytoplasmic delivery of AuNPs. Though adenoviral NLS sequence was incapable of entering the HepG2 cells when included in the cell media (CitationTkachenko et al 2003), it however facilitated transport into the cytoplasm of 3T3/NIH cells and demonstrated some evidence for nuclear translocation in Hela cells (CitationTkachenko et al 2004). It was concluded that the discrepancy observed with the adenoviral NLS peptides may be related to different levels of difficulty in membrane translocation among these three cell lines (CitationTkachenko et al 2003). Nevertheless, AuNPs grafted with a mixture of adenoviral RME and NLS sequence were shown to penetrate the nucleus of HepG2 cells (CitationTkachenko et al 2003). These conjugates even displayed preferential nuclear entry in comparison to the single, long adenoviral peptide that contains both RME and NLS sequence (CitationTkachenko et al 2003; CitationRyan et al 2007). The observed preference was suggested to be due to the spatial accessibility with two shorts sequences providing higher accessibility to the cellular receptors (CitationTkachenko et al 2003).

Similarly, the HIV 1 Tat-protein-derived peptides (CitationLewin et al 2000; CitationFord et al 2001) have facilitated the translocation of AuNPs into the nuclei of human fibroblast cells (Citationde la Furente and Berry 2005). Tat peptide grafted onto the surface of 30 nm AuNPs via tiopronin linker successfully transported AuNPs into the nucleus with no detectable toxicity at concentration up to 10 μM. The cellular transporting pathway of Tat–tiopronin/AuNPs is similar to what was postulated for the NLS–AuNPs conjugates. Tat–AuNPs entered the cytoplasm of human fibroblast cells via RME and translocated into the nucleus through interaction with the nuclear pore.

Unlike the NLS and Tat peptides, most RGD peptides do not induce nuclear translocation. They initiate RME when bound to the RGD receptors overexpressed on the surface of human fibroblast cells. It is however important that the RGD peptide be linked through an appropriate linking moiety as improper conjugation of RGD to the AuNPs has been observed to result in loss of RGD-mediated RME (). For example, a direct coupling of RGD peptides via tiopronin using the same AuNPs platforms described above for the Tat–AuNPs conjugate rendered the RGD–AuNPs conjugates inactive toward human fibroblast cells. This problem could be circumvented by linking RGD peptide onto the tiopronin–AuNPs through secondary linkers, such as ethylenediamine (EDA) and poly(ethylene glycol) bis(3-aminopropyl) terminated (PEG) (CitationMrksich and Whitesides 1996). As expected for a RME-sequenced peptide, RGD–EDA–tiopronin–AuNPs conjugates were internalized within the cytoplasm of the human fibroblast cells. However, no internalization was observed for RGD–PEG–tiopronin–AuNPs conjugates. They remained isolated and adhered to the surface integrin receptors of the human fibroblast cells (Citationde la Furente et al 2006). The discrepancy in RME behavior of these conjugates remained unclear and needs to be investigated further.

Figure 10 Illustration of peptidyl-linker conjugated AuNPs.

Figure 10 Illustration of peptidyl-linker conjugated AuNPs.

Despite the commonly observed lack of nuclear targeting by RGD-derived peptides, nuclear accumulation has been observed in some cell lines, including Hela and HepG2 cell, when incubated with AuNPs grafted with certain uniquely modified RGD peptides (CitationTkachenko et al 2004). The modified RGD peptides consist of sequence from the integrin binding domain in addition to six continuous lysine residues. The nuclear uptake of these modified RGD peptides maybe due to the resemblance of the continuous lysine residues to the lysine-enriched SV40 NLS peptides (CitationTkachenko et al 2004). The low toxicity of these conjugates add to the potential for their use in drug delivery, membrane receptor mapping (Citationde la Furente et al 2006) or even photothermal therapeutic applications.

Small molecule delivery platform

AuNPs have been delivered by and/or facilitated the delivery of assorted small molecule therapeutic agents () into tumors. This delivery is premised on the EPR effect of AuNPs, a property that allows them to be taken up passively (via its leaky vasculature) into tumors without the assistance of targeting agents (CitationSahoo et al 2007). Upon accumulation at the tumor site, the appended small molecule facilitates a RME-mediated uptake of the AuNPs into the diseased cells. One example of such small molecules is folic acid, a form of water soluble vitamin B that has been exploited to selectively target folate receptor (FR) expressing tumor cells (CitationSudimack and Lee 2000; CitationLu and Low 2002; CitationLu et al 2004; CitationRoy et al 2004; CitationStevens et al 2004). FR are overexpressed in various types of human cancers such as the ovary, kidney, breast, brain, lung, prostate, and throat, while generally absent in most normal tissues (CitationSudimack and Lee 2000; CitationLu and Low 2002; CitationBhattacharya et al 2007). AuNPs-folate conjugates have been shown to permit selective targeting of FR-positive tumors. These conjugates have been used in tumor imaging and photothermal therapy applications (CitationDixit et al 2006). Mechanistic studies of internalization of AuNPs–PEG–folate conjugates by KB cells, a human epithelial carcinoma cell line that overexpresses the FR, revealed that the selective uptake of the conjugates is via FR-mediated RME. Dendrimer-entrapped, folate functionalized AuNPs have also shown the potential for targeting and imaging cancer cells (CitationShi et al 2007). Using KB cells that express both high and low levels of FR, these nanoparticles were selectively up taken by the high FR-expressing cells. Subsequent TEM imaging of treated cells revealed a predominant lysosomal localization of the nanoparticles within 2 h of incubation. A similar conjugation of AuNPs with methotrexate (MTX), an analogue of folic acid, has been reported as an alternative formulation strategy to circumvent tumor cell resistance which invariably developed upon repeated use of this versatile anticancer drug (CitationChen et al 2007b). It was shown that MTX–AuNPs conjugates rapidly accumulate in LL2 (Lewis lung carcinoma) cells, inducing higher cytotoxic effects on the tumor compared with free MTX which showed no antitumor effects.

Figure 11 Chemical structures of small molecules functionalized onto the surface of AuNPs.

Figure 11 Chemical structures of small molecules functionalized onto the surface of AuNPs.

AuNPs conjugates of other chemotherapeutic agents have been reported to address various limitations of the unconjugated agents (CitationGanesh 2007; CitationVijayaraghavalu et al 2007). Paciotti and colleagues recently reported a multifunctional vector for targeted drug delivery to solid tumors (CitationPaciotti et al 2006). This vector consists of TNFα, thiolated poly(ethylene glycol) (PT), and paclitaxel (PTX), a leading anticancer drug, which were all bound on the same 26-nm AuNPs. The release of PTX from the vector was investigated in vitro in B16/F10 melanoma tumor cells. It was observed that the vector remained inactive unless treated with dithiothreitol (DDT), suggesting that the vector is acting as a prodrug from which PTX must be released to elicit the desired anticancer effects. In vivo co-administration of cysteamine, an approved therapeutic, with the vector was found to activate PTX release. Compared to unconjugated TNFα and PTX, it was shown that the PTX–PT–AuNPs– TNFα vector delivers 10-fold more TNFα and PTX to the tumor site. A similar system, consisting of two components with different functions: an antiangiogenic molecule, VEGF antibody-2C3 (AbVF), and an anticancer drug, gemcitabine, which are both attached onto a single AuNP core, has been reported (CitationMukherjee et al 2005). Analysis of this conjugate using human umbilical vein endothelial cells (HUVEC) and 786-O cells revealed that the functional integrities of both VEGF antibody and gemcitabine were retained.

AuNPs have also been conjugated with clinically useful antileukemic and antiinflammatory drugs 6-mercaptopurine (6-MP) and its riboside derivative. The resulting conjugates were reported to possess substantially enhanced antiproliferative effects against K-562 leukemia cells compared to the corresponding free forms of these drugs (CitationPodsiadlo et al 2008). In addition, AuNPs–6–MP conjugates have shown antibacterial and antifugal activities against various strains of Gram-positive and Gram-negative organisms including Micrococcus leteus, Staphylococcus aureus, Pseudomonas aeruginosa, Escherichia coli, Aspergillus fumigatus, and Aspergillus niger (CitationSelvaraj et al 2006). This enhanced activity of 6-MP-AuNP conjugate may be attributed to the high penetrating power of AuNPs through the microorganism cell wall, their small size and high surface area. AuNPs conjugates of fluorescent small molecules, such as coumarin, have also been developed as cellular probes and delivery agents (CitationShenoy et al 2006). It was shown that attachment of coumarin, through a carbamate bond, to PEG-functionalized AuNPs caused significant enhancement of emission intensity. Upon incubation with MDA-MB-231 cells, the modified nanoparticles were rapidly internalized in the cells and localized in the perinuclear region as evidenced through intracellular particle tracking.

Most of the AuNPs conjugates reported in the literature have high inorganic (Au metal) contents, necessitating high nanoparticle dosages to elicit the desired effect. The availability of fabrication methods that increase the organic contents of AuNPs will not only lower their dosage-activity ratio, but also extend their utility to other applications where high loading capacity is crucial. Toward this end, CitationGibson and colleagues (2007) recently described a novel approach that permitted high-load functionalization of 2 nm AuNPs with PTX. The resulting hybrid nanoparticles contained a 67 wt % organic content, the highest value reported to date. If proven to be generally applicable, this method may offer an attractive alternative for the preparation of nanosized drug-delivery systems with high drug-loading capacities.

Concluding remarks

In this review, we have focused on the current applications of AuNPs in nanomedicine. We have provided an eclectic collection of AuNPs delivery strategies that are currently under investigation. Assorted classes of vehicles, including small molecules, peptides, and proteins are showing great promise in specific targeting of AuNPs to diseased tissues. In addition, the biocompatibility and photo-optical distinctiveness of AuNPs are now proven to be powerful in diagnostic and biosensing applications, thereby offering a bright hope for the diagnosis and treatment of many disease states.

The sustained fascination of the scientific community with AuNPs research have been facilitated by significant strides in many fronts including availability of a plethora of methods for the production and functionalization of AuNPs of various shapes and sizes. It is now possible to control particle sizes at nanometer resolution. Improved understanding of molecular targeting in biology has furnished several ligands that have been successfully used for specific delivery of AuNPs. With information accruing from proteomics studies on various diseases, one expects that many more ligands will be made available for AuNPs-targeted delivery.

However, the successful implementations of the promised applications of AuNPs are still limited in part by the formidable barriers imposed by the complexity of a whole organism in contrast to simple cell based studies that formed the bed rock of most of the proof-of-principle investigations. The recent results from the phase I clinical trial on Aurimune™, indicating a safe and targeted delivery of Aurimune™ in and around tumor sites (CitationCytImmune 2008), are particularly intriguing and encouraging. This has provided very important evidence that AuNPs-based therapeutic agents could overcome the barriers presented by the human immune and circulatory systems to achieve delivery at diseased sites without uptake by healthy tissues. In principle, such improved targeted delivery could make other AuNPs-based experimental therapeutic techniques, such as photothermal therapy, practicable. With the “right” combination of delivery agents and particle size, AuNPs-based therapeutics could effectively kill the diseased cells while eliminating the horrendous side effects of the conventional chemotherapeutic agents. Nevertheless, more still needs to be done regarding our understanding of the pharmacokinetics and toxicity profiles of AuNPs. Special attention should be given to gaining comprehensive insights on the effects of nanoparticle size, ligand conjugation and conjugation chemistry on AuNPs physiological properties. Additionally, the potential for cumulative toxicity upon repeated exposure to AuNPs-based agents must be rigorously investigated. Nanotoxicity may not be a small matter after all! Results from these and related studies will prove informative in further refinement of the design of AuNPs for use in various nanotechnology applications.

Acknowledgments

This work was financially supported by Georgia Institute of Technology and by the Blanchard fellowship to AK Oyelere. P Chen is a recipient of the GAANN predoctoral fellowship from the Georgia Tech Center for Drug Design, Development and Delivery.

References

  • AgarwalAHuangSWO’DonnellM2007Targeted gold nanorod contrast agent for prostate cancer detection by photoacoustic imagingJ Appl Phys1020647011
  • AhmedSMSalgiaR2006Epidermal growth factor receptor mutations and susceptibility to targeted therapy in lung cancerRespirology116879217052295
  • AimeSBottaMFasanoM1998Lanthanide(III) chelates for NMR biomedical applicationsChem Soc Rev2719
  • AkermanMEChanWCWLaakkonenP2002Nanocrystal targeting in vivoProc Natl Acad Sci U S A99126172112235356
  • AllenTMHansenCMartinF1991Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivoBiochim Biophys Acta106629362065067
  • ArteagaCL2001The epidermal growth factor receptor: from mutant oncogene in nonhuman cancers to terapeutic target in human neoplasiaJ Clin Oncol1932s40s11560969
  • AubinMEMoralesDGHamad-SchifferliK2005Labeling ribonuclease S with a 3 nm Au nanoparticle by two-step assemblyNano Lett55192215755106
  • BakóJSzepesiMMártonI2007Synthesis of nanoparticles for dental drug delivery systemsFogorvosi Szemle1001091317695047
  • BallouBLagerholmBCErnstLA2004Noninvasive imaging of quantum dots in miceBioconjug Chem15798614733586
  • BaronRWillnerBWillnerI2007Biomolecule–nanoparticle hybrids as functional units for nanobiotechnologyChem Commun2832332
  • BernardiRJLoweryARThompsonPA2008Immunonanoshells for targeted photothermal ablation in medulloblastoma and glioma: an in vitro evaluation using human cell linesJ Neurooncol861657217805488
  • BhattacharyaRPatraCREarlA2007Attaching folic acid on gold nanoparticles using noncovalent interaction via different polyethylene glycol backbones and targeting of cancer cellsNanomedicine322438
  • BoyerDTamaratPMaaliA2002Photothermal imaging of nanometer-sized metal particles among scatterersScience2971160312183624
  • BritoLAmijiM2007Nanoparticulate carriers for the treatment of coronary restenosisInt J Nanomedicine21436117722543
  • BrouckaertPGLeroux-RoelsGGGuisezY1986In vivo antitumour activity of recombinant human and murine TNF, alone and in combination with murine IFN-, on a syngeneic murine melanomaInt J Cancer3876393095251
  • BrownKRNatanMJ1998Hydroxylamine seeding of colloidal Au nanoparticles in solution and on surfacesLangmuir147268
  • BruchezMJrMoronneMGinP1998Semiconductor nanocrystals as fluorescent biological labelsScience281201369748157
  • BusbeeBDObareSOMurphyCJ2003An improved synthesis of high-aspect-ratio gold nanorodsJ Adv Mater154146
  • CaoYCJinRMirkinCA2002Nanoparticles with raman spectroscopic fingerprints for DNA and RNA detectionScience29715364012202825
  • CaravanPEllisonJJMcMurryTJ1999Gadolinium(III) chelates as MRI contrast agents: Structure, dynamics, and applicationsChem Rev99229335211749483
  • CaravanPGreenwoodJMWelchJT2003Gadolinium-binding helix–turn–helix peptides: DNA-dependent MRI contrast agentsChem Commun2025745
  • CarusoRAAntoniettiM2001Sol-gel nanocoating: An approach to the preparation of structured materialsChem Mater13327282
  • ChahSHammondMRZareRN2005Gold nanoparticles as a colorimetric sensor for protein conformational changesChem Biol12323815797216
  • ChanWCMaxwellDJGaoX2002Luminescent quantum dots for multiplexed biological detection and imagingCurr Opin Biotechnol1340611849956
  • ChenJSaekiFWileyBJ2005Gold nanocages: bioconjugation and their potential use as optical imaging contrast agentsNano Lett5473715755097
  • ChenJWangDXiJ2007aImmuno gold nanocages with tailored optical properties for targeted photothermal destruction of cancer cellsNano Lett713182217430005
  • ChenYTsaiCHuangP2007bMethotrexate conjugated to gold nanoparticles inhibits tumor growth in a syngeneic lung tumor modelMol Pharm47132217708653
  • ChengMMCudaGBunimovichYL2006Nanotechnologies for biomolecular detection and medical diagnosticsCurr Opin Chem Biol1011916418011
  • ChoKWangXNieS2008Therapeutic nanoparticles for drug delivery in cancerClin Cancer Res141310618316549
  • CognetLTardinCBoyerD2003Single metallic nanoparticle imaging for protein detection in cellsProc Natl Acad Sci U S A10011350513679586
  • CohenuramMSaifMW2007Epidermal growth factor receptor inhibition strategies in pancreatic cancer: past, present and the futureJ Pancreas8415
  • ConnorEEMwamukaJGoleA2005Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicitySmall1325717193451
  • CytImmune2008Aurimune™ (CYT-6091) [online]Accessed on Sept 1, 2008URL: http://www.cytimmune.com/go.cfm?do=Page.View&pid&=26
  • de la FurenteJMBerryCCRiehleMO2006Nanoparticle targeting at cellsLangmuir2232869316548590
  • de la FurenteJMBerryCC2005Tat peptide as an efficient molecule to translocate gold nanoparticles into the cell nucleusBioconjug Chem1611768016173795
  • DeMYouC-CSrivastavaS2007Biomimetic interactions of proteins with functionalized nanoparticles: a thermodynamic studyJ Am Chem Soc129107475317672456
  • DebouttiereP-JRouxSVocansonF2006Design of gold nanoparticles for magnetic resonance imagingAdv Funct Mater1623309
  • DemannETSteinPSHaubenreichJE2005Gold as an implant in medicine and dentistryJ Long Term Eff Med Implants156879816393135
  • DixitVVan den BosscheJShermanDM2006Synthesis and grafting of thioctic acid-PEG-folate conjugates onto Au nanoparticles for selective targeting of folate receptor-positive tumor cellsBioconjug Chem17603916704197
  • DongSRomanM2007Fluorescently labeled cellulose nanocrystals for bioimaging applicationsJ Am Chem Soc12913810117949004
  • DurrNJLarsonTSmithDK2007Two-photon luminescence imaging of cancer cells using molecularly targeted gold nanorodsNano Lett7941517335272
  • EdelmanERSeifertPGroothuisA2001Gold-coated NIR stents in porcine coronary arteriesCirculation1034293411157696
  • ElghanianRStorhoffJJMucicRC1997Selective colorimetric detection of polynucleotides based on the distance-dependent optical properties of gold nanoparticlesScience2771078819262471
  • El-SayedIHHuangXEl-SayedMA2005aSelective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticlesCancer Lett2391293516198049
  • El-SayedIHHuangXEl-SayedMA2005bSurface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics: applications in oral cancerNano Lett58293415884879
  • EvertsMSainiVLeddonJL2006Covalently linked Au nanoparticles to a viral vector: potential for combined photothermal and gene cancer therapyNano Lett65879116608249
  • FeldherrMCAkinDJ1990The permeability of the nuclear envelope in dividing and nondividing cell culturesJ Cell Biol111182365731
  • FordKGSouberbielleBEDarlingD2001Protein transduction: an alternative to genetic intervention?Gene Ther81411402295
  • FrederixFFriedtJ-MChoiK-H2003Biosensing based on light absorption of nanoscaled gold and silver particlesAnal Chem75689490014670050
  • FreitasRAJr2005What is nanomedicine?Nanomedicine12917292052
  • FrensG1973Controlled nucleation for the regulation of the particle size in monodisperse gold suspensionsNature Phys Sci241202
  • GaneshT2007Improved biochemical strategies for targeted delivery of taxoidsBioorg Med Chem15359762317419065
  • GaoXCuiYLevensonRM2005In vivo cancer targeting and imaging with semiconductor quantum dotsNat Biotechnol229697615258594
  • GhoshmoulickRBhattacharyaJMitraCK2007Protein seeding of gold nanoparticles and mechanism of glycation sensingNanomedicine32081417692576
  • GibsonJDKhanalBPZubarevER2007Paclitaxel-functionalized gold nanoparticlesJ Am Chem Soc129116536117718495
  • GielenMTiekinkERT2005Metallotherapeutic Drugs and Metal-Based Diagnostic Agents: The Use of Metals in MedicineHoboken, NJJohn Wiley and Sons
  • GoodmanCMMcCuskerCDYilmazT2004Toxicity of gold nanoparticles functionalized with cationic and anionic side chainsBioconjug Chem1589790015264879
  • HaTHYeongJYChungBH2005Immobilization of hexa-arginine tagged esterase onto carboxylated gold nanoparticlesChem Commun31395961
  • HabaYKojimaCHaradaA2007Preparation of poly(ethylene glycol)-modified poly(amido amine) dendrimers encapsulating gold nanoparticles and their heat-generating abilityLangmuir235243617419657
  • HainfeldJFSlatkinDNFocellaTM2006Gold nanoparticles: a new X-ray contrast agentJ Radiol7924853
  • HallerCHizohI2004The cytotoxicity of iodinated radiocontrast agents on renal cells in vitroInvest Radiol391495415076007
  • HanGGhoshPDeM2007Drug and gene delivery using gold nanoparticlesNanoBioTech3405
  • HanYJiangJLeeSS2008Reverse microemulsion-mediated synthesis of silica-coated gold and silver nanoparticlesLangmuir245842818465888
  • HayatM1989Colloidal Gold Principles, Methods, and ApplicationsSan Diego, CAAcademic Press
  • HeathJRDavisME2008Nanotechnology and cancerAnnu Rev Med592516517937588
  • HerrwerthSEckWReinhardtS2003Factors that determine the protein resistance of oligoether self-assembled monolayers – internal hydrophilicity, terminal hydrophilicity, and lateral packing densityJ Am Chem Soc12593596612889964
  • HieberUHeimME1994Tumor necrosis factor for the treatment of malignanciesOncology51142538196898
  • HillRTShearJB2006Enzyme-nanoparticle functionalization of three-dimensional protein scaffoldsAnal Chem787022617007529
  • HirschLRStaffordRJBanksonJA2003Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidanceProc Natl Acad Sci U S A100135495414597719
  • HizohIHallerC2002Radiocontrast-induced renal tubular cell apoptosis: hypertonic versus oxidative stressInvest Radiol374283412138358
  • HuaizhiZYuantaoN2001China’s ancient gold drugsGold Bull34249
  • HuangC-CYangZChangH-T2004Synthesis of dumbbell-shaped Au-Ag core-shell nanorods by seed-mediated growth under alkaline conditionsLangmuir2060899215248687
  • HuangXEl-SayedIHQianW2006Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorodsJ Am Chem Soc12821152016464114
  • HuangXJainPKEl-SayedIH2007aGold nanoparticles: interesting optical properties and recent applications in cancer diagnostics and therapyNanomed268193
  • HuangXJainPKEl-SayedIH2008Plasmonic photothermal therapy (PPTT) using gold nanoparticlesLasers Med Sci232172817674122
  • HuangXQianWEl-SayedIH2007bThe potential use of the enhanced nonlinear properties of gold nanospheres in photothermal cancer therapyLasers Surg Med397475317960762
  • HudsonBIHofmannMABucciarelliL2002Glycation and diabetes: The RAGE connectionCurr Sci83151521
  • IpeBIYoosafKThomasKG2006Functionalized gold nanoparticles as phosphorescent nanomaterials and sensorsJ Am Chem Soc12819071316464092
  • IshidaOMaruyamaKSasakiK1999Size-dependent extravasation and interstitial localization of polyethyleneglycol liposomes in solid tumor-bearing miceInt J Pharm190495610528096
  • JainKK2008Nanomedicine: application of nanobiotechnology in medical practiceMed Princ Pract178910118287791
  • JainPKLeeKSEl-SayedIH2006Calculated absorption and scattering properties of gold nanoparticles of different size, shape, and composition: applications in biological imaging and biomedicineJ Phys Chem B11072384816599493
  • JanaNRGearheartLMurphyCJ2001aSeed-mediated growth approach for shape-controlled synthesis of spheroidal and rodlike gold nanoparticles using a surfactant templateJ Adv Mater13138993
  • JanaNRGearheartLMurphyCJ2001bWet chemical synthesis of high aspect ratio cylindrical gold nanorodsJ Phys Chem B10540657
  • JenaBKRajCR2006Electrochemical biosensor based on integrated assembly of dehydrogenase enzymes and gold nanoparticlesAnal Chem786332916970306
  • JinRCCaoYWMirkinCA2001Photoinduced conversion of silver nanospheres to nanoprismsScience2941901311729310
  • KalderonDRichardsonWDMarkhamAF1984Sequence requirements for nuclear location of simian virus 40 large-T antigenNature (London)3113386088992
  • KawasakiESPlayerA2005Nanotechnology, nanomedicine, and the development of new, effective therapies for cancerNanomedicine1101917292064
  • KhlebtsovNGTrachukLAMel’nikovAG2005The effect of the size, shape, and structure of metal nanoparticles on the dependence of their optical properties on the refractive index of a disperse mediumOpt Spectrosc988390
  • KimDParkSLeeJH2007Antibiofouling polymer-coated gold nanoparticles as a contrast agent for in vivo X-ray computed tomography imagingJ Am Chem Soc1297661517530850
  • KimDKMikhaylovaMWangFH2003Starch-coated superparamagnetic nanoparticles as MR contrast agentsChem Mater15434351
  • KimSLimYTSolteszEG2004Near-infrared fluorescent type II quantum dots for sentinel lymph node mappingNat Biotechnol2293714661026
  • KircheisROstermannEWolschekMF2002Tumor-targeted gene 1 of tumor necrosis factor-alpha induces tumor necrosis and tumor regression without systemic toxicityCancer Gene Ther96738012136428
  • KohlerNFryxellGEZhangM2004A bifunctional poly(ethylene glycol) silane immobilized on metallic oxide-based nanoparticles for conjugation with cell targeting agentsJ Am Chem Soc12672061115186157
  • KooOMRubinsteinIOnyukselH2005Role of nanotechnology in targeted drug delivery and imaging: a concise reviewNanomedicine119321217292079
  • KreibigUGenzelL1985Optical absorption of small metallic particlesSurf Sci156678700
  • KumarCSSR2007Nanomaterials for Cancer DiagnosisWeinheim, GermanyWiley-VCH
  • KunduSPanigrahiSPraharajS2007Anisotropic growth of gold clusters to gold nanocubes under UV irradiationNanotechnology1807571221730520
  • KunduSPengLLiangH2008A new route to obtain high-yield multiple-shaped gold nanoparticles in aqueous solution using microwave radiationInorg Chem4763445218563880
  • LangereisSKooistraHAvan GenderenMH2004Probing the interaction of the biotin – avidin complex with the relaxivity of biotinylated Gd-DTPAOrg Biomol Chem21271315105914
  • LanzaGWinterPCyrusT2006Nanomedicine opportunities in cardiologyAnn N Y Acad Sci10804516517132801
  • LeeHLeeEKimDK2006aAntibiofouling polymer-coated superparamagnetic iron oxide nanoparticles as potential magnetic resonance contrast agents for in vivo cancer imagingJ Am Chem Soc1287383916734494
  • LeeJHHuhMJunYW2006bArtificially engineered magnetic nanoparticles for ultra-sensitive molecular imagingNat Med1395917187073
  • LeeK-SEl-SayedMA2006Gold and silver nanoparticles in sensing and imaging: sensitivity of plasmon response to size, shape, and metal compositionJ Phys Chem B11019220517004772
  • LeeLAWangQ2006Adaptations of nanoscale viruses and other protein cages for medical applicationsNanomedicine21374917292136
  • LewinMCarlessoNTungC-H2000Tat peptide-derivatized magnetic nanoparticles allow in vivo tracking and recovery of progenitor cellsNat Biotechnol18410410748521
  • LewinskiNColvinVDrezekR2008Cytotoxicity of nanoparticlesSmall4264918165959
  • LewisDJDayTMMacPhersonJV2006Luminescent nanobeads: attachment of surface reactive Eu(III) complexes to gold nanoparticlesChem Commun1314335
  • LiHRothbergL2004Colorimetric detection of DNA sequences based on electrostatic interactions with unmodified gold nanoparticlesProc Natl Acad Sci U S A10114036915381774
  • LiuYShiptonMKKaufmanED2007Synthesis, stability, and cellular internalization of gold nanoparticles containing mixed peptide-poly(ethylene glycol) monolayersAnal Chem792221917288407
  • LooCLoweryAHalasN2005Immunotargeted nanoshells for integrated cancer imaging and therapyNano Lett57091115826113
  • LoweryARGobinAMDayES2006Immunonanoshells for targeted photothermal ablation of tumor cellsInt J Nanomedicine11495417722530
  • LuC-WHungYHsiaoJ-K2007Bifunctional magnetic silica nanoparticles for highly efficient human stem cell labelingNano Lett71495417212455
  • LuLWangHZhouY2002Seed-mediated growth of large, monodisperse core – shell gold – silver nanoparticles with Ag-like optical propertiesChem Commun21445
  • LuYLowPS2002Folate-mediated delivery of macromolecular anticancer therapeutic agentsAdv Drug Deliv Rev546759312204598
  • LuYSegaELeamonCP2004Folate receptor-targeted immunotherapy of cancer: mechanism and therapeutic potentialAdv Drug Deliv Rev5611617615094213
  • LynchTJBellDWSordellaR2004Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinibN Engl J Med35021293915118073
  • MaedaHWuJSawaT2000Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a reviewJ Control Release652718410699287
  • MahmoodUWeisslederR2003Near-infrared optical imaging of proteases in cancerMol Cancer Ther24899612748311
  • MajorosIJMycAThomasT2006PAMAM dendrimer-based multifunctional conjugate for cancer therapy: synthesis, characterization, and functionalityBiomacromolecules7572916471932
  • MarinakosSMNovakJPBrousseauLC1999Gold particles as templates for the synthesis of hollow polymer capsules. control of capsule dimensions and guest encapsulationJ Am Chem Soc121851822
  • MartinaMSFortinJPMenagerC2005Generation of superparamagnetic liposomes revealed as highly efficient MRI contrast agents for in vivo imagingJ Am Chem Soc127106768516045355
  • MaysingerD2007Nanoparticles and cells: good companions and doomedrelationshipsOrg Biomol Chem523354217637950
  • McFaddenP2002Broadband biodetection: Holmes on a chipScience2972075612242448
  • MerbachATothE2001The Chemistry of Contrast Agent in Magnetic Resonance ImagingChichester, UKWiley
  • MitraRNDasPK2008In situ preparation of gold nanoparticles of varying shape in molecular hydrogel of peptide amphiphilesJ Phys Chem C112815966
  • MoolhuizenGPaciottiGFde LeedeLGJ2004Colloidal gold nanoparticlesLondon, UKBusiness Briefing, Pharmatech
  • MorrowKJJBawaRWeiC2007Recent advances in basic and clinical nanomedicineMed Clin North Am918054317826104
  • MrksichMWhitesidesGM1996Using self-assembled monolayers to understand the interactions of man-made surfaces with proteins and cellsAnnu Rev Biophys Biomol Struct2555788800464
  • MuLFengSS2003A novel controlled release formulation for the anticancer drug paclitaxel (Taxol®): PLGA nanoparticles containing vitamin E TPGSJ Control Release86334812490371
  • MukherjeePBhattacharyaRMukhopadhyayD2005Gold nanoparticles bearing functional anti-cancer drug and anti-angiogenic agent: A “2 in 1” system with potential application in cancer therapeuticsJ Biomed Nanotech12248
  • MurphyCJJanaNR2002Controlling the aspect ratio of inorganic nanorods and nanowiresAdv Mater14802
  • NasongklaNBeyERenJ2006Multifunctional polymeric micelles as cancer-targeted, MRI-ultrasensitive drug delivery systemsNano Lett624273017090068
  • [NCI] National Cancer Institute2003Computed tomography (CT): Questions and answers [online]Accessed on Sept 1, 2008URL: http://www.cancer.gov/cancertopics/factsheet/Detection/CT
  • NicholasJDLarsonTSmithDK2007Two-photon luminescence imaging of cancer cells using molecularly targeted gold nanorodsNano Lett7941517335272
  • NiidomeTYamagataMOkamotoY2006PEG-modified gold nanorods with a stealth character for in vivo applicationsJ Control Release114343716876898
  • NorthRJHavellEA1988The antitumor function of tumor necrosis factor (TNF) II. Analysis of the role of endogenous TNF in endotoxin-induced hemorrhagic necrosis and regression of an established sarcomaJ Exp Med1671086993258350
  • OkazakiK-IKiyamaTHiraharaK2008Single-step synthesis of gold – silver alloy nanoparticles in ionic liquids by a sputter deposition techniqueChem Commun66913
  • OtsukaHAkiyamaYNagasakiY2001Quantitative and reversible lectin-induced association of gold nanoparticles modified with -lactosyl – mercapto-poly(ethylene glycol)J Am Chem Soc12382263011516273
  • OyelereAKChenPCHuangX2007Pepetide-conjugated gold nanorods for nuclear targetingBioconjug Chem181490717630680
  • PaciottiGFKingstonDGITamarkinL2006Colloidal gold nanoparticles: a novel nanoparticle platform for developing multifunctional tumor-targeted drug delivery vectorsDrug Dev Res674754
  • PaciottiGFMyerLWeireichD2004Colloidal gold: a novel nanoparticle vector for tumor directed drug deliveryDrug Deliv111698315204636
  • PaezJPJannePALeeJC2004EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapyScience304149750015118125
  • PalAEsumiK2007Photochemical synthesis of biopolymer coated Aucore-Agshell type bimetallic nanoparticlesJ Nanosci Nanotechnol72110517655002
  • PalSDeG2007Synthesis of Au-Ag alloy nanoparticles with Au/Ag compositional control in SiO2 film matrixJ Nanosci Nanotechnol71994917654979
  • PanYNeussSLeifertA2007Size-dependent cytotoxicity of gold nanoparticlesSmall31941917963284
  • PandeyPSinghSPAryaSK2007Application of thiolated gold nanoparticles for the enhancement of glucose oxidase activityLangmuir233333717261046
  • PapahadjopoulosDAllenTMGabizonA1991Sterically stablized liposomes: improvements in pharmacokinetics and antitumor therapeutic efficacyProc Natl Acad Sci U S A881146041763060
  • PetersR2006Nanoscopic medicine: The next frontierSmall2452617193067
  • PissuwanDValenzuelaSMMillerCM2007A golden bullet? Selective targeting of toxoplasma gondii tachyzoites using antibody-functionalized gold nanorodsNano Lett738081218034505
  • PodsiadloPSinaniVABahngJH2008Gold nanoparticles enhance the anti-leukemia action of a 6-mercaptopurine chemotherapeutic agentLangmuir245687418052300
  • PratoMKostarelosKBiancoA2008Functionalized carbon nanotubes in drug design and discoveryAcc Chem Res4160817867649
  • PrudkinLWistubaII2006Epidermal growth factor receptor abnomalities in lung cancer. Pathogentic and clinical implicationsAnn Diagn Pathol103061516979526
  • PulliamBSungJCEdwardsDA2007Design of nanoparticle-based dry powder pulmonary vaccinesExpert Opin Drug Deliv46516317970667
  • PyrpassopoulosSNiarchosDNouneisG2007Synthesis and self-organization of Au nanoparticlesNanotechnology18485604
  • QianXPengX-HAnsariDO2008In vivo tumor targeting and spectroscopic detection with surface-enhanced Raman nanoparticle tagsNat Biotechnol26839018157119
  • RaghunuandNGatenbyRAGilliesRJ2003Microenvironmental and cellular consequences of altered blood flow in tumoursBr J Radiol76S11S2215456710
  • RagusaAGarcíaIPenadésS2007Nanoparticles as nonviral gene delivery vectorsIEEE Trans Nanobioscience63193018217625
  • RawatMSinghDSarafS2006Nanocarriers: promising vehicle for bioactive drugsBiol Pharm Bull291790816946487
  • RaynalIPrigentPPeyramaureS2004Macrophage endocytosis of superparamagnetic iron oxide nanoparticles: mechanisms and comparison of ferumoxides and ferumoxtran-10Invest Radiol39566314701989
  • RichardsDGMcMIllinDLMeinEA2002Gold and its relationship to neurological/glandular conditionsInt J Neurosci112315312152404
  • RogerWJBasuP2005Factors regulating macrophage endocytosis of nanoparticles: implications for targeted magnetic resonance plaque imagingAtherosclerosis178677315585202
  • RoyEJGawlickUOrrBA2004Folate-mediated targeting of T cells to tumorsAdv Drug Deliv Rev5612193115094217
  • RyanJAOvertonKWSpeightME2007Cellular uptake of gold nanoparticles passivated with BSA-SV40 large t antigen conjugatesAnal Chem799150917973401
  • SahooSKLabhasetwarV2003Nanotech approaches to drug delivery and imagingDrug Discov Today811122014678737
  • SahooSKParveenSPandaJJ2007The present and future of nanotechnology in human health careNanomedicine3203117379166
  • SarkarDHalasNJ1997General vector basis function solution of Maxwell’s equationsPhys Rev E561102
  • SatoKHosokawaKMaedaM2003Rapid aggregation of gold nanoparticles induced by non-cross-linking DNA hybridizationJ Am Chem Soc1258102312837070
  • SatoKHosokawaKMaedaM2005Non-cross-linking gold nanoparticle aggregation as a detection method for single-base substitutionsNucleic Acids Res33e415640441
  • SauTKMurphyCJ2004Room temperature, high-yield synthesis of multiple shapes of gold nanoparticles in aqueous solutionJ Am Chem Soc1268648915250706
  • SelvarajVAlagarMHamertonI2006Analytical detection and biological assay of antileukemic drug using gold nanoparticlesElectrochim Acta52115260
  • SharmaPBrownSGWalter2006Nanoparticles for bioimagingAdv Colloid Interface Sci123647185
  • ShawIII FC1999Gold-based therapeutic agentsChem Rev99258960011749494
  • ShenoyDFuWLiJ2006Surface functionalization of gold nanoparticles using hetero-bifunctional poly(ethylene glycol) spacer for intracellular tracking and deliveryInt J Nanomedicine151716467923
  • ShiXWangSMeshinchIS2007Dendrimer-entrapped gold nanoparticles as a platform for cancer-cell targeting and imagingSmall312455217523182
  • ShuklaRBansalVChaudharyM2005Biocompatibility of gold nanoparticles and their endocytotic fate inside the cellular compartment: a microscopic overviewLangmuir21106445416262332
  • SimonianALGoodTAWangS-S2005Nanoparticle-based optical biosensors for the direct detection of organophosphate chemical warfare agents and pesticidesAnal Chim Acta5346977
  • SkrabalakSEAuLLuX2007Gold nanocages for cancer detection and treatmentNanomed265768
  • SonnichsenCFranzlTWilkT2002Drastic reduction of plasmon damping in gold nanorodsPhys Rev Lett8807740211863939
  • SrinivasanJMFajardoLFHahnGM1990Mechanism of antitumor activity of tumor necrosis factor alpha with hyperthermia in a tumor necrosis factor alpha-resistant tumorJ Natl Cancer Inst821904102250311
  • StevensPJSekidoMLeeRJ2004A folate receptor-targeted lipid nanoparticle formulation for a lipophilic paclitaxel prodrugPharm Res212153715648245
  • StonehuernerJGZhaoJO’DalyJP1992Comparison of colloidal gold electrode fabrication methods: the preparation of a horseradish peroxidase enzyme electrodeBiosens Bioelectron742181515118
  • StreicherRMSchmidtMFioritoS2007Nanosurfaces and nanostructures for artificial orthopedic implantsNanomed286174
  • SudimackJLeeRJ2000Targeted drug delivery via the folate receptorAdv Drug Deliv Rev411476210699311
  • SunRWMaDLWongEL2007Some uses of transition metal complexes as anti-cancer and anti-HIV agentsDalton Trans4348849217992273
  • SunYMayersBXiaY2003Metal nanostructures with hollow interiorsAdv Mater156416
  • SunYXiaY2003Gold and silver nanoparticles: A class of chromo-phores with colors tunable in the range from 400 to 750 nmAnalyst1286869112866889
  • SvedmanCDunérKKehlerM2006Lichenoid reactions to gold from dental restorations and exposure to gold through intracoronary implant of a gold-plated stentClin Res Cardiol956899117066351
  • SvedmanCTillmanCGustavssonCG2005Contact allergy to gold in patients with gold-plated intracoronary stentsContact Dermatitis52192615859991
  • ThelenABauknechtHCAsbachP2006Behavior of metal implants used in ENT surgery in 7 Tesla magnetic resonance imagingEur Arch Otorhinolaryngol263900516835741
  • ThurnKTBrownEMBWuA2007Nanoparticles for applications in cellular imagingNanoscale Res Lett24304121794189
  • TkachenkoAGXieHColemanD2003Multifunctional gold nanoparticle-peptide complexes for nuclear targetingJ Am Chem Soc1254700112696875
  • TkachenkoAGXieHColemanD2004Cellular trajectories of peptide-modified gold particle complexes: comparison of nuclear localization signals and peptide transduction domainsBioconjug Chem154829015149175
  • TorchilinVPLukyanovANGaoZ2003Immunomicelles: Targeted pharmaceutical carriers for poorly soluble drugsProc Natl Acad Sci U S A10060394412716967
  • TsaiCSYuTBChenCT2005Gold nanoparticle-based competitive colorimetric assay for detection of protein–protein interactionsChem Commun3442735
  • TurkevichJStevensonPCHillierJ1951A study of the nucleation and growth process in the synthesis of colloidal goldDiscuss Faraday Soc115575
  • UmenoHWatanabeHYamauchiN1994Enhancement of blood stasis and vascular permeability in Meth-A tumors by administration of hyperthermia in combination with tumor necrosis factorJpn J Cancer Res85325308188532
  • VijayaraghavaluSRaghavanDLabhasetwarV2007Nanoparticles for delivery of chemotherapeutic agents to tumorsCurr Opin Investig Drugs847784
  • VillalongaRCaoRGragosoA2007Supramolecular chemistry of cyclodextrin in enzyme technologyChem Rev107308811617590054
  • VisariaRKGriffinRJWilliamsBW2006Enhancement of tumor thermal therapy using gold nanoparticle-assisted tumor necrosis factor-alpha deliveryMol Cancer Ther510142016648573
  • WangHHuffTBZweifelDA2005In vitro and in vivo two photon luminescence imaging of single gold nanorodsProc Natl Acad Sci U S A10215752616239346
  • WeisslederRMahmoodU2001Molecular imagingRadiology2193163311323453
  • WoodleMCEngbersCMZalipskyS1994New amphipatic polymer-lipid conjugates forming long-circulating reticuloendothelial system-evading liposomesBioconjug Chem549367873652
  • XiongHQAbbruzzeseIL2002Epidermal growth factor receptor-targeted therapy for pancreatic cancerSemin Oncol2931712422311
  • YangPHSunXChiuJF2005Transferrin-mediated gold nanoparticle cellular uptakeBioconjug Chem16494615898713
  • YehHCHoYPWangTH2005Quantum dot – mediated biosensing assays for specific nucleic acid detectionNanomedicine11152117292066
  • YguerabideJYguerabideEE1998Light-scattering submicroscopic particles as highly fluorescent analogs and their use as tracer labels in clinical and biological applications II. experimental characterizationAnal Biochem262157769750129
  • ZhaoWJeffreyCFLChiumanW2008Enzymatic cleavage of nucleic acids on gold nanoparticles: a generic platforms for facile colorimetric biosensorsSmall4810618537135
  • ZharovVPKimJWCurielDT2005Self-assembling nanoclusters in living systems: application for integrated photothermal nanodiagnostics and nanotherapyNanomedicine13264517292107
  • ZhengMDavidsonFHuangX2003Ethylene glycol monolayer protected nanoparticles for eliminating nonspecific binding with biological moleculesJ Am Chem Soc1257790112822983