Singh 2018
Singh 2018
Singh 2018
Molecular Sciences
Review
Gold Nanoparticles in Diagnostics and Therapeutics
for Human Cancer
Priyanka Singh 1 , Santosh Pandit 2 , V.R.S.S. Mokkapati 2 , Abhroop Garg 1 ID
,
Vaishnavi Ravikumar 1 and Ivan Mijakovic 1,2, *
1 The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark,
2800 Kgs. Lyngby, Denmark; [email protected] (P.S.); [email protected] (A.G.);
[email protected] (V.R.)
2 Systems and Synthetic Biology Division, Department of Biology and Biological Engineering,
Chalmers University of Technology, 41296 Chalmers, Sweden; [email protected] (S.P.);
[email protected] (V.R.S.S.M.)
* Correspondence: [email protected]; Tel.: +46-070-982-8446
Received: 8 June 2018; Accepted: 3 July 2018; Published: 6 July 2018
Abstract: The application of nanotechnology for the treatment of cancer is mostly based on
early tumor detection and diagnosis by nanodevices capable of selective targeting and delivery
of chemotherapeutic drugs to the specific tumor site. Due to the remarkable properties of gold
nanoparticles, they have long been considered as a potential tool for diagnosis of various cancers
and for drug delivery applications. These properties include high surface area to volume ratio,
surface plasmon resonance, surface chemistry and multi-functionalization, facile synthesis, and stable
nature. Moreover, the non-toxic and non-immunogenic nature of gold nanoparticles and the high
permeability and retention effect provide additional benefits by enabling easy penetration and
accumulation of drugs at the tumor sites. Various innovative approaches with gold nanoparticles are
under development. In this review, we provide an overview of recent progress made in the application
of gold nanoparticles in the treatment of cancer by tumor detection, drug delivery, imaging,
photothermal and photodynamic therapy and their current limitations in terms of bioavailability and
the fate of the nanoparticles.
Keywords: gold nanoparticles; cancer; protein corona; biocompatibility; photoimaging; drug delivery;
photothermal therapy; photodynamic therapy; clinical trials; toxicology
1. Introduction
With recent advances in nanotechnology and medical science, numerous nanoparticles and
nanomaterials have emerged from different bulk elements such as gold, silver, iron, copper, cobalt,
platinum, etc., which are synthesized either biologically or physiochemically [1,2]. The ability to
manipulate nanoparticle features, such as their physical, chemical and biological properties, opens up
many possibilities to explore these nanoparticles in drug delivery as image contrast agents and for
diagnostic purposes [3]. Among various organic and inorganic nanoparticles, gold nanoparticles
possess unique optical and Surface Plasmon Resonance (SPR) properties, due to which it has become
the first choice for researchers, particularly in the biological and pharmaceutical field (Figure 1). Due to
the optical properties of gold nanoparticles, they are especially utilized in ultrasensitive detection
and imaging-based therapeutic techniques required for the treatment of lethal diseases, such as
cancer. Cancer is a disease state caused by abnormal cell growth and is the third leading cause of
mortality worldwide. According to the World Health Organization (WHO, www.who.int), cancer
caused 8.8 million deaths in 2015. Current cancer treatment is based on chemotherapeutic drugs,
usually involving chemo or radiation therapy, with the aim to kill the cancer cells [4,5]. However,
these treatments often result in several side effects due to the damage caused to the surrounding
healthy tissues. In addition, delays in diagnosis and a high incidence of relapse result in lower survival
rates. Treating cancer cells by utilizing a nanoparticle-based drug delivery approach plays a key role
in overcoming the limitations of conventional treatment methodologies by providing simultaneous
diagnosis and treatment [4]. Consequently, a considerable amount of research focusing on gold
nanoparticle-based nanocarrier development and their potential applications in cancer biology and
nanomedicine has been carried out [6]. In this review, we focus on providing further new insights for
exploring the gold nanoparticle applications as a tool in cancer diagnostics and treatment.
Typically, gold nanoparticles of controlled size and shape have been synthesized by various
physical (microwave and ultraviolet (UV) irradiation, laser ablation), chemical, and biological ways.
Chemical synthesis generally utilizes chemicals and solvents, which are associated to environmental
and human health impacts. In addition, it demands extreme conditions (e.g., pH, temperature) which
are not optimal [7–9]. On the other hand, biological nanoparticle synthesis (plants and microorganisms
mediated) is a relatively new, eco-friendly, and promising area of research with a considerable
potential for expansion [10–12]. Numerous medicinal plants have shown potential to produce stable
gold nanoparticles within a few seconds [11,13–15]. Microorganisms are also equally capable of
adsorbing gold atoms and accumulating gold nanoparticles by secreting large amounts of enzymes,
which are involved in the enzymatic reduction of gold ions [16,17]. These biologically synthesized
gold nanoparticles have become an attractive and potential option to explore as a tool for biosensors,
immunoassays, targeted drug delivery, photoimaging, photothermal therapy (PTT), and photodynamic
therapy (PDT) (Figure 2). Interestingly, in human cancer and cell biology, various types of gold
nanoparticles, such as gold nanorods, nanocages, nanostars, nanocubes, and nanospheres, have become
effective tools. Their application in cancer diagnostics and therapeutic development is due to their
favorable optical and physical properties that provide a potential platform for developing cancer
theranostics. The optical properties of gold nanoparticles rely on SPR. In principal, SPR is a process
whereby the electrons of gold resonate in response to an incoming radiation, causing them to both
absorb and scatter light. In addition, some specifically shaped gold nanoparticles contribute to photon
capture cross sections that are four to five-fold greater than those of photothermal dyes. These attributes
Int. J. Mol. Sci. 2018, 19, 1979 3 of 16
are exploited to obtain localized heating either to destroy the cells or for drug release, underlying the
therapeutic applications. In addition, gold nanoparticles possess tunable properties, which allow for
the synthesis of nanoparticles of specific size and desired shape, resulting in a plasmonic resonance shift
from 520 to 800–1200 nm (complex shapes) [18]. Susie et al. showed the change in optical properties
and resonance of gold nanoparticles (ranging from 500 to 1200) by slightly changing the nanoparticles’
shape from nanospheres of 15–30 nm to nanorods of 2.5–7.5 Aspect ratio (AR.) [19] The range between
800 and 1200 is therapeutically useful because the body tissue is moderately transparent to Near
Infra-Red (NIR) light, thereby providing an opportunity for therapeutic effects in deep tissues by
photothermal and photoimaging approaches. Another important property is the available surface
area. It is well known that the surface area of nanoparticles is inversely proportional to their size,
which results in a large surface area to volume ratio. In other words, nanoparticle have a large surface
area available for drug loading, conjugation, or binding of any gene or biological moiety of choice,
thus increasing drug solubility, stability, and pharmacokinetic parameters [20]. The available surface
area also plays a critical role for the application of gold nanoparticles in cancer diagnostics, specifically
in photo-imaging and photothermal therapy. In photothermal therapy, smaller nanoparticles are
preferred as light is mainly adsorbed by the nanoparticles and thus efficiently converted to heat for
destruction of cell, whereas in photo-imaging, lager nanoparticles are preferred because of their higher
scattering efficiency. In addition, biological responses to nanoparticles tend to scale with surface area.
This means that when nanoparticles are exposed to a biological environment, such as serum or plasma,
more proteins from the surroundings bind to small nanoparticles with a larger surface area-to-volume
ratio than to those with a larger size and a smaller surface area-to-volume ratio. In parallel with
the above-mentioned properties, the tailored surface functionalization of gold nanoparticles has
also evinced considerable interest. The possibility to conjugate gold nanoparticles with a variety of
biologically active moieties, especially with amine and thiol groups, provides possibilities for important
biomedical applications ranging from diagnostics, targeting specific delivery of drugs/genes, imaging,
and sensing for electron microscopy markers [21,22].
Despite all these benefits, biocompatibility of gold nanoparticles is a crucial factor to take into
account prior to clinical applications. Although the inert nature of gold nanoparticles makes them
relatively biocompatible, the cytotoxicity of the nanoparticles, which is more or less dependent on
Int. J. Mol. Sci. 2018, 19, 1979 4 of 16
their shape, size, surface properties, and chemical composition, has to be further evaluated. Once the
particles are internalized by the cells, the proteins present in the physiological environment form
a coating called a “corona” on the surface of the nanoparticles, resulting in a nanoparticle-protein
complex [23]. This protein corona is quite complex and variable in structure and plays a key role in the
biodistribution of nanoparticles throughout the body. Dobrovolskaia et al. reported that untreated
plasma usually contains approx. 3700 proteins, and the gold nanoparticles that come in contact with the
plasma form a “protein corona complex” containing fewer than 100 proteins [24]. Gold nanoparticles
inside the corona complex contain opsonins on their surface, which are recognized by the immune
cells (part of reticuloendothelial system (RES)). These proteins ultimately determine the route of
nanoparticle internalization and eventually affect the fate of the nanoparticles in the body (i.e., rate and
route of clearance from the bloodstream and body, volume of distribution, organ disposition, etc.)
(Figure 3) [25]. So far, various proteins have been reported to have been isolated from the corona
complex in the plasma, such as albumin, fibrinogen, Immunoglobulin G (IgG), Immunoglobulin M
(IgM), transferrin, etc. This corona complex can cause changes in particle size and charge, which in
turn affect the internalization process into the macrophages of the RES and the overall distribution
in the body. Certain proteins allow macrophages to easily recognize nanoparticles; for example,
IgG opsonins, and fibrinogens are reported to promote phagocytosis and nanoparticles removal from
the body [26], whereas dysopsonins such as albumins are reported to cause prolonged blood circulation
of nanoparticles [27]. To prevent the nanoparticles from immune recognition, scientists introduced
a process called “PEGylation”. In this approach, nanoparticles “hide” by masking their surface with
a poly-ethyleneglycol (PEG) layer. This saves them from immune recognition, in essence prolonging
their blood circulation. PEGylation can be done by covalent linking that entraps or adsorbs PEG
chains onto the surface of the nanoparticle. Once the nanoparticles are internalized, they can be
used in tumor imaging, PTT, and PDT. Although PEGylation can help in avoiding rapid recognition
by the RES, complete avoidance is rarely achieved as nanoparticles may still be recognized and
taken up by the RES system (Figure 4). Though a lot of work in this regard has been conducted,
many challenges remain and must be tackled before PEGylation can be put into practice. In this review,
we summarize the importance and advantages of gold nanoparticles and their utilization in several
aspects of cancer therapeutics.
Figure 3. Distribution of nanoparticles with varying coatings and bound proteins. PEG = poly-ethyleneglycol.
Int. J. Mol. Sci. 2018, 19, 1979 5 of 16
Figure 4. Systemic delivery of multifunctional gold nanoparticles for cancer bioimaging, photothermal
therapy (PPT), and photodynamic therapy (PDT). EPR = enhanced permeation and retention;
NIR = near infra-red; ROS = reactive oxygen species.
their use as drug carriers [29,30]. For example, methotrexate (MTX), which has been used to treat cancer
for decades, upon conjugation with gold nanoparticles displayed higher cytotoxicity towards numerous
tumor cell lines as compared to that of free MTX. MTX was observed to accumulate in the tumor cells
at a faster rate and to a higher level when conjugated with gold nanoparticles [31]. Another drug,
doxorubicin (DOX), when bound to gold nanoparticles via an acid labile linker, showed enhanced
toxicity against the multi drug resistant MCF-7/ADR breast cancer cell line, thus overcoming the multi
drug resistance to some extent due to the enhanced uptake of the gold nanoparticle-tethered drug
followed by its responsive release within the cell [32]. In the past, peptide-drug-conjugates (PDCs) have
been investigated for their use as anticancer agents [33–36]. However, their stability in the blood, liver,
and kidneys pose a significant challenge to their successful use as an anticancer molecule. Recently,
it was shown that this difficulty can be by-passed by conjugating these PDCs to gold nanoparticles.
The authors reported an increase in the half-life of PDCs from 10.6–15.4 min (administered alone),
to 21.0–22.3 h (upon conjugation with gold nanoparticle), while retaining cytotoxicity [37]. Apart from
synthetic drugs, phytochemicals have also shown the potential of being used as anticancer drugs but,
similar to PDCs, they too have certain problems such as low specificity, short half-life, fast clearance
rate, and inefficient cell penetration. These problems in using phytochemicals can be by-passed by
conjugating them to gold nanoparticles. For example, kaempferol (a phytochemical) conjugated to
gold nanoparticles displayed both significantly higher apoptosis and inhibition of angiogenesis in
MCF-7 breast cancer cells as compared to kaempferol alone [38]. Table 1 lists some of the studies
performed to investigate the anti-tumor applications of gold nanoparticles in drug delivery.
Nanoparticle
Nanoparticle Outcome Cell Lines Ref.
Size (nm)
Higher cytotoxicity towards numerous cell
lines as compared to free MTX. Lewis lung carcinoma
MTX-AuNP 8–80 [31]
Suppression of tumor growth with (LL2) cells
MTX-AuNP but not with free MTX.
Enhanced toxicity against multi drug
DOX-Hyd@AuNP 30 MCF-7/ADR cancer cells [32]
resistant cancer cells.
A549 lung epithelial
Platinum-tethering exhibited higher
cancer cell line, HCT116,
(Pt(R,R-dach))-AuNP 26.7 cytotoxicity as compared to free oxaliplatin [39]
HCT15, HT29, and RKO
that could enter the nucleus.
colon cancer cell lines
Cellular uptake of targeted particles was
Tfpep-AuNP conjugated with LN229 and U87 human
5.1 significantly higher than that of the [40]
photodynamic pro-drug Pc 4 glioma cancer lines
non-targeted ones.
Higher cell death as compared to
CPP-DOX-AuNP 25 HeLa cells and A549 cells [41]
previously tested 41 nm AuNP.
Enhanced drug accumulation and retention
HepG2-R, C0045C,
FA-Au-SMCC-DOX as compared to free DOX in multi drug [42]
and HDF
resistant cancer cells.
Increased efficacy of BHC against
FA-BHC-AuNP 20–60 Vero and HeLa [43]
cancer cells.
Enhanced cellular uptake and cytotoxicity 4T1 mouse mammary
Au-P(LA-DOX)-b-PEG-OH/FA NP 34 [44]
against cancer cells. carcinoma cell line
Induction of early and late apoptosis in
A549, H460, and H520
DOX@PVP-AuNP 12 lung cancer cells and upregulation of tumor [45]
human lung cancer cells
suppression genes.
Enhanced half-maximal effective drug
DOX-BLM-PEG-AuNP 10 concentration, providing rationale for HeLa cells [46]
chemotherapy using two drugs.
The biomimetic nanoparticle loaded with
4T1 mouse mammary
EpCam-RPAuN 48 PTX was used in combination treatment [47]
carcinoma cell line
(PTT and chemotherapy).
AuNP: Gold nanoparticle, AuN: Gold nanocage, BHC: Berberine hydrochloride, BLM: Bleomycin, CPP:
Cell penetrating peptides, DOX: Doxorubicin, EpCam: epithelial cell adhesion molecule, FA: Folic acid, Hyd:
Hydrazone, MTX: Methotrexate, PEG: Poly ethylene glycol, PLA: Poly L-aspartate, (Pt(R,R-dach)): Active ingredient
of oxaliplatin, PTT: Photothermal therapy, PTX: Paclitaxel, PVP: Polyvinylpyrrolidone, SMCC: Succinimidyl
4-(N-maleimidomethyl) cyclohexane-1-carboxylate, Tfpep: Transferrin peptide.
Int. J. Mol. Sci. 2018, 19, 1979 7 of 16
has to be noted here is the effect of PTT therapy on PDT or vice versa. Liu et al. presented a detailed
analysis of this dual therapy where they clearly showed that the hypoxic environment induced by PDT
does not affect PTT as PTT is an oxygen-independent therapy.
Photoimaging is an advanced technique that can help detect early stage tumors and guide the
surgeons for precision treatment. One of the biggest challenges today for the surgeons is to have a clear
picture of where the tumor ends and the healthy tissue begins. During an operation, the surgeons
face a nearly impossible task of deciding to what extent the tumor has to be removed: being too
conservative can leave some tumor cells behind, and being too liberal can result in removing healthy
tissues that can be vital. Being too conservative is the reason that most of the tumors recur with time.
Magnetic Resonance Imaging (MRIs) and Computed Tomography (CT) scans are limited and can
only detect tumors above a size of several millimeters or approximately 10 million cells, meaning
that the tumors are detected only when they reach a certain threshold. Photoimaging is a novel
approach in cancer treatment where millions of functionalized gold nanoparticles are site specifically
injected into the tumor, where they specifically bind to the cancer cells and scatter (shine), making
it easier for the surgeons to identify the tumor and healthy cells. Gold nanoparticles (nanorods,
nanocages, and nanoshells) are known to be the best available photo imaging nanoparticles for cancer
therapeutics due to their bio inertness and their ability to provide increased spacial and temporal
resolution for imaging [65].
the safety evaluation of NU-0129, a spherical nucleic acid (SNA) formulation composed of small
interfering RNAs (siRNAs) targeting the Bcl-2-like protein 12 (BCL2L12) sequence and conjugated
to gold nanoparticles, that has potential antineoplastic activity (NCT03020017). Kharlamov et al.
studied the safety and feasibility of two delivery techniques for gold nanoparticles for the treatment of
atherosclerosis. The first is a PTT approach with silica-gold nanoparticles and the second is a magnetic
navigation approach with silica-gold iron bearing nanoparticles (NCT01270139) [74]. Results obtained
in this trial suggested that PTT using gold-silica nanoparticles is associated with a significant regression
of coronary atherosclerosis and an acceptable level of safety for clinical practice. CNM-Au8 is a gold
nanocrystal suspension drug developed by Clene Nanomedicine (Salt Lake City, UT, USA) for the
demyelinating disorder neuromyelitis optica (NMO) (NCT02755870). A randomized placebo controlled
trial is ongoing in healthy individuals to evaluate the safety, tolerability, and pharmacokinetics of
CNM-Au8. Another gold-nanoparticle based clinical trial is currently underway, with the aim to
evaluate the feasibility of a novel method in oncology involving breath analysis with a nanosensor
array for identifying gastric diseases (NCT01420588). It has been suggested that the nanosensor
array could provide the missing non-invasive screening tool to distinguish gastric cancer and related
precancerous lesions [75]. A clinical trial on another novel diagnostic approach (electronic nose sensor)
with gold nanoparticles is ongoing for the evaluation of its performance in the diagnosis of pulmonary
arterial hypertension (NCT02782026).
Clinical trials.gov
Name Materials Application
Identifier
Laser responsive thermal ablation of solid
Silica-gold nanoshells coated NCT00848042,
AuroLase® tumors: head/neck cancer, primary and/or
with PEG NCT01679470
metastatic lung tumors
Prostate, head and neck, lung MRI/US fusion
Silica-gold nanoshells coated imaging and biopsy in combination with
AuroLase® NCT02680535
with PEG nanoparticle-directed focal therapy for ablation
of prostate tissue
A Spherical Nucleic Acid Targeting BCL2L12 in recurrent glioblastoma
NU-0129 NCT03020017
(SNA) Gold Nanoparticle multiforme or gliosarcoma patients
Silica-Gold Plasmonic photothermal therapy of
Silica-Gold Nanoparticles NCT01270139
Nanoparticles flow-limiting atherosclerotic lesions
Evaluation of safety, tolerability, and
CNM-Au8 gold nanocrystal pharmacokinetics of CNM-Au8 in healthy male NCT02755870
and female volunteers
Sensors functionalized with gold nanoparticles
Gold Nanoparticles Gold nanoparticles Organic functionalized gold nanoparticles NCT01420588
Detection of gastric lesions
Exhaled breath olfactory signature of
Gold Nanoparticles Gold nanoparticles NCT02782026
pulmonary arterial hypertension
5. Current Limitations
As described above, gold nanoparticles do show promise and potential to be used in cancer
diagnostics and therapeutics. Nevertheless, it is imperative to consider the other side of the coin,
i.e., unintended side effects on human health. A number of individual studies previously addressed
the cytotoxicity, effect of size on toxicity, efficacy, biodistribution, retention time, and physiological
response of nanoparticles. However, many of them are seen to contradict one another. Absence of
coherent information on the actual effect of nanoparticles could have delirious effects and a negative
impact on human health. While the discussed issues in general are applicable to any nanoparticle,
examples described below are specific to gold nanoparticles.
Toxicity: The toxicity of gold nanoparticles to biological systems has always been an issue of
concern. Properties of gold nanoparticles such as shape, size, surface chemistry, targeting ligand,
elasticity, and composition largely influence their toxicity. This, in combination with the complexity and
the heterogeneity that exists amongst human cells and tissues, makes it challenging to comprehensively
Int. J. Mol. Sci. 2018, 19, 1979 10 of 16
probe the effect and response of the biological system to the administration of gold nanoparticles.
Surface charge has been reported to influence toxicity of gold nanoparticles, wherein positively
charged particles were found to be more toxic than negative or neutral particles [76]. On the other
hand, other groups found no toxicity induced by positively charged gold nanoparticles [77] and no
toxicity of negatively charged particles [78]. This discrepancy arises due to the unique physiochemical
nature of nanoparticles, and no single standardized assay is currently available that could universally
be applied to test the toxicity effect of all nanoparticles. The lack of such robust standardized assays
leads to varying interpretations or assumptions that limit nanoparticle administration. Toxicity assay
using Caenorhabditis elegans (ISO 10872 method) is widely used to assess the effect of nanoparticles on
multicellular organisms. Hanna et al. recently reported an artifact caused while testing the toxicity of
positively charged gold nanoparticles using the C. elegans assay [79]. The authors initially observed
growth inhibition of the nematodes when fed with E. coli along with a suspension of positively
charged gold nanoparticles. However, they deduced that this observation was a false positive as
gold nanoparticles heteroagglomerate with E. coli cells, influencing the ability of the nematodes to
feed. On repeating the assay in the absence of E. coli, the authors observed a reduced toxicity effect,
illustrating unforeseen artifacts that could occur in such widely used toxicity assays. Ginzburg et al.
showed a synergistic toxicity effect induced by gold nanoparticles in the presence of additives such
as surfactants, while the individual components separately exhibited low toxicity [80], underlining
the importance of identifying strategies for selecting safe nanoparticles-additive/ligand/modifier
combinations. Gold nanoparticles were also shown to have species-specific differences with respect
to biodistribution, pathophysiologic response, and retention time. Bahamonde et al. observed that
gold nanoparticle-treated mice and rats responded differently, wherein a number of rats died on gold
nanoparticle administration while no fatality was seen amongst the mice [81]. Additionally, the authors
also noticed a relatively higher accumulation of the gold nanoparticle in rats as compared to the mice,
highlighting the fact of differential physiological response even amongst closely related groups.
Size and Biodistribution: Apart from toxicity assessment, size and biodistribution of nanoparticles
are also significant factors to take into consideration. Tang et al. reported increased cytotoxicity of
smaller gold nanoparticles (8 nm) coated with reduced glutathione when tested on a human hepatic cell
line as compared to that of the larger particles (37 nm) [82]. On the other hand, Rosli et al. recorded that
50 nm gold nanoparticles exhibited higher cytotoxicity in a breast cancer cell line as compared to their
13 and 70 nm counterparts [83]. Connor et al. studied the cytotoxicity of a series of gold nanoparticle
sizes ranging from 4 to 18 nm on human leukemia cells and found that none of the sizes were not
harmful to cellular function [84]. Liang et al. showed that PEG-coated gold nanoparticles of 4.8 nm had
the highest toxicity effect on Hela cells whereas the 12.1 and 27.3 nm counterparts showed low toxicity
and the 46.6 nm counterpart exhibited absolutely no toxicity [85]. Li et al., however, reported that
regardless of the size of the nanoparticles, the observed cytotoxicity was due to dose-dependency [86].
Sonavane et al. noticed a similar accumulation of gold nanoparticles in the liver while testing the
effect of nanoparticle size on biodistribution. The authors observed that gold nanoparticles of all
sizes mainly accumulated in organs like liver, lung, and spleen. The 15 nm particles accumulated in
tissues including blood, liver, lung, spleen, kidney, brain, heart, and stomach whereas much larger
particles (200 nm) showed a very minute presence in organs including blood, brain, stomach, and
pancreas [87]. With respect to biodistribution, Fraga et al. assessed the biodistribution of ~20 nm
citrate- and pentapeptide CALNN (cysteine–alanine–leucine–asparagine–asparagine) -coated gold
nanoparticles and found them to mainly accumulate in liver [88]. Cho et al. analyzed gold contents
of 13 nm PEG-coated gold nanoparticles and found them to accumulate in the liver and spleen [89].
Li et al. showed the accumulation of larger PEG-coated gold nanoparticles (42.5 and 61.2 nm) in the
liver. Additionally, the authors also observed a longer retention time (poor elimination rate) possibly
causing further safety issues [86]. Aside from those issues mentioned above, it is vital to take into
account the non-biodegradability and non-porous properties of gold nanoparticles which would likely
have an effect on the pharmacokinetics [90].
Int. J. Mol. Sci. 2018, 19, 1979 11 of 16
Thus, currently, there is controversy and inconsistency regarding the potential of gold
nanoparticles for clinical applications, and there is an inherent need for the development of universally
applicable methods to evaluate the biocompatibility of gold nanoparticles and to have a firm
understanding of their interaction with the living system. Lastly, it is also worthwhile to reflect on
whether the cost of synthesis would justify the underlying therapeutic capabilities of gold nanoparticles
(concept discussed in general in [91]).
Acknowledgments: We acknowledge the financial support from the H.C. Ørsted fellowship, co-funded by
Marie Skłodowska Curie, to P.S. and Novo Nordisk Foundation and VINNOVA to I.M.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Singh, H.; Du, J.; Singh, P.; Mavlonov, G.T.; Yi, T.H. Development of superparamagnetic iron oxide
nanoparticles via direct conjugation with ginsenosides and its in-vitro study. J. Photochem. Photobiol. B Biol.
2018, 185, 100–110. [CrossRef] [PubMed]
2. Schrofel, A.; Kratosova, G.; Safarik, I.; Safarikova, M.; Raska, I.; Shor, L.M. Applications of biosynthesized
metallic nanoparticles—A review. Acta Biomater. 2014, 10, 4023–4042. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2018, 19, 1979 12 of 16
3. Tao, Y.; Li, M.; Ren, J.; Qu, X. Metal nanoclusters: Novel probes for diagnostic and therapeutic applications.
Chem. Soc. Rev. 2015, 44, 8636–8663. [CrossRef] [PubMed]
4. Singh, P.; Kim, Y.J.; Singh, H.; Ahn, S.; Castro-Aceituno, V.; Yang, D.C. In situ preparation of water-soluble
ginsenoside Rh2-entrapped bovine serum albumin nanoparticles: In vitro cytocompatibility studies. Int. J. Nanomed.
2017, 12, 4073–4084. [CrossRef] [PubMed]
5. Singh, P.; Singh, H.; Castro-Aceituno, V.; Ahn, S.; Kim, Y.J.; Farh, M.E.-A.; Yang, D.C. Engineering of
mesoporous silica nanoparticles for release of ginsenoside CK and Rh2 to enhance their anticancer and
anti-inflammatory efficacy: In vitro studies. J. Nanopart. Res. 2017, 19, 257. [CrossRef]
6. Pissuwan, D.; Valenzuela, S.M.; Cortie, M.B. Therapeutic possibilities of plasmonically heated gold nanoparticles.
Trends Biotechnol. 2006, 24, 62–67. [CrossRef] [PubMed]
7. Singh, P.; Ahn, S.; Kang, J.P.; Veronika, S.; Huo, Y.; Singh, H.; Chokkaligam, M.; El-Agamy Farh, M.;
Aceituno, V.C.; Kim, Y.J.; et al. In vitro anti-inflammatory activity of spherical silver nanoparticles and
monodisperse hexagonal gold nanoparticles by fruit extract of Prunus serrulata: A green synthetic approach.
Artif. Cells Nanomed. Biotechnol. 2017, 1–11. [CrossRef] [PubMed]
8. Singh, H.; Du, J.; Yi, T.H. Kinneretia THG-SQI4 mediated biosynthesis of silver nanoparticles and its
antimicrobial efficacy. Artif. Cells Nanomed. Biotechnol. 2017, 45, 602–608. [CrossRef] [PubMed]
9. Singh, H.; Du, J.; Yi, T.H. Biosynthesis of silver nanoparticles using Aeromonas sp. THG-FG1.2 and its antibacterial
activity against pathogenic microbes. Artif. Cells Nanomed. Biotechnol. 2017, 45, 584–590. [CrossRef] [PubMed]
10. Singh, P.; Kim, Y.J.; Zhang, D.; Yang, D.C. Biological Synthesis of Nanoparticles from Plants and Microorganisms.
Trends Biotechnol. 2016, 34, 588–599. [CrossRef] [PubMed]
11. Singh, H.; Du, J.; Yi, T.H. Green and rapid synthesis of silver nanoparticles using Borago officinalis leaf extract:
Anticancer and antibacterial activities. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1310–1316. [CrossRef] [PubMed]
12. Singh, P.; Kim, Y.J.; Singh, H.; Wang, C.; Hwang, K.H.; Farh Mel, A.; Yang, D.C. Biosynthesis, characterization,
and antimicrobial applications of silver nanoparticles. Int. J. Nanomed. 2015, 10, 2567–2577.
13. Singh, P.; Kim, Y.J.; Yang, D.C. A strategic approach for rapid synthesis of gold and silver nanoparticles by
Panax ginseng leaves. Artif. Cells Nanomed. Biotechnol. 2016, 44, 1949–1957. [CrossRef] [PubMed]
14. Singh, P.; Kim, Y.J.; Wang, C.; Mathiyalagan, R.; El-Agamy Farh, M.; Yang, D.C. Biogenic silver and gold
nanoparticles synthesized using red ginseng root extract, and their applications. Artif. Cells Nanomed. Biotechnol.
2016, 44, 811–816. [CrossRef] [PubMed]
15. Singh, H.; Du, J.; Singh, P.; Yi, T.H. Ecofriendly synthesis of silver and gold nanoparticles by Euphrasia officinalis leaf
extract and its biomedical applications. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1163–1170. [CrossRef] [PubMed]
16. Singh, P.; Singh, H.; Kim, Y.J.; Mathiyalagan, R.; Wang, C.; Yang, D.C. Extracellular synthesis of silver and
gold nanoparticles by Sporosarcina koreensis DC4 and their biological applications. Enzym. Microb. Technol.
2016, 86, 75–83. [CrossRef] [PubMed]
17. Singh, P.; Kim, Y.J.; Singh, H.; Mathiyalagan, R.; Wang, C.; Yang, D.C. Biosynthesis of Anisotropic
Silver Nanoparticles by Bhargavaea indica and Their Synergistic Effect with Antibiotics against Pathogenic
Microorganisms. J. Nanomater. 2015, 2015, 234741. [CrossRef]
18. Aldewachi, H.; Chalati, T.; Woodroofe, M.N.; Bricklebank, N.; Sharrack, B.; Gardiner, P. Gold nanoparticle-based
colorimetric biosensors. Nanoscale 2017, 10, 18–33. [CrossRef] [PubMed]
19. Eustis, S.; El-Sayed, M.A. Why gold nanoparticles are more precious than pretty gold: Noble metal surface
plasmon resonance and its enhancement of the radiative and nonradiative properties of nanocrystals of
different shapes. Chem. Soc. Rev. 2006, 35, 209–217. [CrossRef] [PubMed]
20. Kumar, D.; Saini, N.; Jain, N.; Sareen, R.; Pandit, V. Gold nanoparticles: An era in bionanotechnology. Expert Opin.
Drug Deliv. 2013, 10, 397–409. [CrossRef] [PubMed]
21. Sanvicens, N.; Marco, M.P. Multifunctional nanoparticles–properties and prospects for their use in human
medicine. Trends Biotechnol. 2008, 26, 425–433. [CrossRef] [PubMed]
22. Zhang, Q.; Yang, M.; Zhu, Y.; Mao, C. Metallic Nanoclusters for Cancer Imaging and Therapy. Curr. Med. Chem.
2018, 25, 1379–1396. [CrossRef] [PubMed]
23. Aggarwal, P.; Hall, J.B.; McLeland, C.B.; Dobrovolskaia, M.A.; McNeil, S.E. Nanoparticle interaction with
plasma proteins as it relates to particle biodistribution, biocompatibility and therapeutic efficacy. Adv. Drug
Deliv. Rev. 2009, 61, 428–437. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2018, 19, 1979 13 of 16
24. Dobrovolskaia, M.A.; Patri, A.K.; Zheng, J.; Clogston, J.D.; Ayub, N.; Aggarwal, P.; Neun, B.W.; Hall, J.B.;
McNeil, S.E. Interaction of colloidal gold nanoparticles with human blood: Effects on particle size and
analysis of plasma protein binding profiles. Nanomed. Nanotechnol. Biol. Med. 2009, 5, 106–117. [CrossRef]
[PubMed]
25. Chen, D.; Ganesh, S.; Wang, W.; Amiji, M. Plasma protein adsorption and biological identity of systemically
administered nanoparticles. Nanomedicine 2017, 12, 2113–2135. [CrossRef] [PubMed]
26. Goppert, T.M.; Muller, R.H. Adsorption kinetics of plasma proteins on solid lipid nanoparticles for drug
targeting. Int. J. Pharm. 2005, 302, 172–186. [CrossRef] [PubMed]
27. Ogawara, K.; Furumoto, K.; Nagayama, S.; Minato, K.; Higaki, K.; Kai, T.; Kimura, T. Pre-coating with serum
albumin reduces receptor-mediated hepatic disposition of polystyrene nanosphere: Implications for rational
design of nanoparticles. J. Control. Release 2004, 100, 451–455. [CrossRef] [PubMed]
28. Bahrami, B.; Hojjat-Farsangi, M.; Mohammadi, H.; Anvari, E.; Ghalamfarsa, G.; Yousefi, M.; Jadidi-Niaragh, F.
Nanoparticles and targeted drug delivery in cancer therapy. Immunol. Lett. 2017, 190, 64–83. [CrossRef]
[PubMed]
29. Ajnai, G.; Chiu, A.; Kan, T.; Cheng, C.C.; Tsai, T.H.; Chang, J. Trends of Gold Nanoparticle-based Drug
Delivery System in Cancer Therapy. J. Exp. Clin. Med. 2014, 6, 172–178. [CrossRef]
30. Kong, F.Y.; Zhang, J.W.; Li, R.F.; Wang, Z.X.; Wang, W.J.; Wang, W. Unique Roles of Gold Nanoparticles in
Drug Delivery, Targeting and Imaging Applications. Molecules 2017, 22, 1445. [CrossRef] [PubMed]
31. Chen, Y.H.; Tsai, C.Y.; Huang, P.Y.; Chang, M.Y.; Cheng, P.C.; Chou, C.H.; Chen, D.H.; Wang, C.R.; Shiau, A.L.;
Wu, C.L. Methotrexate conjugated to gold nanoparticles inhibits tumor growth in a syngeneic lung tumor
model. Mol. Pharm. 2007, 4, 713–722. [CrossRef] [PubMed]
32. Wang, F.; Wang, Y.C.; Dou, S.; Xiong, M.H.; Sun, T.M.; Wang, J. Doxorubicin-tethered responsive gold
nanoparticles facilitate intracellular drug delivery for overcoming multidrug resistance in cancer cells.
ACS Nano 2011, 5, 3679–3692. [CrossRef] [PubMed]
33. Firer, M.A.; Laptev, R.; Kasatkin, I.; Trombka, D. Specific destruction of hybridoma cells by antigen-toxin
conjugates demonstrate an efficient strategy for targeted drug therapy in leukemias of the B cell lineage.
Leuk. Lymphoma 2003, 44, 681–689. [CrossRef] [PubMed]
34. Gellerman, G.; Baskin, S.; Galia, L.; Gilad, Y.; Firer, M.A. Drug resistance to chlorambucil in murine B-cell
leukemic cells is overcome by its conjugation to a targeting peptide. Anticancer Drugs 2013, 24, 112–119.
[CrossRef] [PubMed]
35. Gilad, Y.; Firer, M.A.; Rozovsky, A.; Ragozin, E.; Redko, B.; Albeck, A.; Gellerman, G. “Switch off/switch on”
regulation of drug cytotoxicity by conjugation to a cell targeting peptide. Eur. J. Med. Chem. 2014, 85, 139–146.
[CrossRef] [PubMed]
36. Gilad, Y.; Noy, E.; Senderowitz, H.; Albeck, A.; Firer, M.A.; Gellerman, G. Dual-drug RGD conjugates
provide enhanced cytotoxicity to melanoma and non-small lung cancer cells. Biopolymers 2015, 106, 160–171.
[CrossRef] [PubMed]
37. Kalimuthu, K.; Lubin, B.C.; Bazylevich, A.; Gellerman, G.; Shpilberg, O.; Luboshits, G.; Firer, M.A.
Gold nanoparticles stabilize peptide-drug-conjugates for sustained targeted drug delivery to cancer cells.
J. Nanobiotechnol. 2018, 16, 34. [CrossRef] [PubMed]
38. Srinivas Raghavan, B.; Kondath, S.; Anantanarayanan, R.; Rajaram, R. Kaempferol mediated synthesis of
gold nanoparticles and their cytotoxic effects on MCF-7 cancer cell line. Process Biochem. 2015, 50, 1966–1976.
[CrossRef]
39. Brown, S.D.; Nativo, P.; Smith, J.A.; Stirling, D.; Edwards, P.R.; Venugopal, B.; Flint, D.J.; Plumb, J.A.;
Graham, D.; Wheate, N.J. Gold nanoparticles for the improved anticancer drug delivery of the active
component of oxaliplatin. J. Am. Chem. Soc. 2010, 132, 4678–4684. [CrossRef] [PubMed]
40. Dixit, S.; Novak, T.; Miller, K.; Zhu, Y.; Kenney, M.E.; Broome, A.M. Transferrin receptor-targeted theranostic
gold nanoparticles for photosensitizer delivery in brain tumors. Nanoscale 2015, 7, 1782–1790. [CrossRef]
[PubMed]
41. Park, H.; Tsutsumi, H.; Mihara, H. Cell-selective intracellular drug delivery using doxorubicin and alpha-helical
peptides conjugated to gold nanoparticles. Biomaterials 2014, 35, 3480–3487. [CrossRef] [PubMed]
42. Cheng, J.; Gu, Y.-J.; Cheng, S.H.; Wong, W.-T. Surface Functionalized Gold Nanoparticles for Drug Delivery.
J. Biomed. Nanotechnol. 2013, 9, 1362–1369. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2018, 19, 1979 14 of 16
43. Pandey, S.; Mewada, A.; Thakur, M.; Shah, R.; Oza, G.; Sharon, M. Biogenic gold nanoparticles as fotillas to
fire berberine hydrochloride using folic acid as molecular road map. Mater. Sci. Eng. C Mater. Biol. Appl.
2013, 33, 3716–3722. [CrossRef] [PubMed]
44. Prabaharan, M.; Grailer, J.J.; Pilla, S.; Steeber, D.A.; Gong, S. Gold nanoparticles with a monolayer of
doxorubicin-conjugated amphiphilic block copolymer for tumor-targeted drug delivery. Biomaterials 2009,
30, 6065–6075. [CrossRef] [PubMed]
45. Ramalingam, V.; Varunkumar, K.; Ravikumar, V.; Rajaram, R. Target delivery of doxorubicin tethered with
PVP stabilized gold nanoparticles for effective treatment of lung cancer. Sci. Rep. 2018, 8, 3815. [CrossRef]
[PubMed]
46. Farooq, M.U.; Novosad, V.; Rozhkova, E.A.; Wali, H.; Ali, A.; Fateh, A.A.; Neogi, P.B.; Neogi, A.; Wang, Z.
Gold Nanoparticles-enabled Efficient Dual Delivery of Anticancer Therapeutics to HeLa Cells. Sci. Rep. 2018,
8, 2907. [CrossRef] [PubMed]
47. Zhu, D.M.; Xie, W.; Xiao, Y.S.; Suo, M.; Zan, M.H.; Liao, Q.Q.; Hu, X.J.; Chen, L.B.; Chen, B.; Wu, W.T.;
et al. Erythrocyte membrane-coated gold nanocages for targeted photothermal and chemical cancer therapy.
Nanotechnology 2018, 29, 084002. [CrossRef] [PubMed]
48. Dreaden, E.C.; Austin, L.A.; Mackey, M.A.; El-Sayed, M.A. Size matters: Gold nanoparticles in targeted
cancer drug delivery. Ther. Deliv. 2012, 3, 457–478. [CrossRef] [PubMed]
49. Kim, C.K.; Ghosh, P.; Pagliuca, C.; Zhu, Z.J.; Menichetti, S.; Rotello, V.M. Entrapment of hydrophobic drugs
in nanoparticle monolayers with efficient release into cancer cells. J. Am. Chem. Soc. 2009, 131, 1360–1361.
[CrossRef] [PubMed]
50. Weissleder, R. A clearer vision for in vivo imaging. Nat. Biotechnol. 2001, 19, 316–317. [CrossRef] [PubMed]
51. Huff, T.B.; Tong, L.; Zhao, Y.; Hansen, M.N.; Cheng, J.X.; Wei, A. Hyperthermic effects of gold nanorods on
tumor cells. Nanomedicine 2007, 2, 125–132. [CrossRef] [PubMed]
52. El-Sayed, I.H.; Huang, X.; El-Sayed, M.A. Selective laser photo-thermal therapy of epithelial carcinoma using
anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett. 2006, 239, 129–135. [CrossRef] [PubMed]
53. Van de Broek, B.; Devoogdt, N.; D’Hollander, A.; Gijs, H.L.; Jans, K.; Lagae, L.; Muyldermans, S.; Maes, G.;
Borghs, G. Specific cell targeting with nanobody conjugated branched gold nanoparticles for photothermal
therapy. ACS Nano 2011, 5, 4319–4328. [CrossRef] [PubMed]
54. Pitsillides, C.M.; Joe, E.K.; Wei, X.; Anderson, R.R.; Lin, C.P. Selective cell targeting with light-absorbing
microparticles and nanoparticles. Biophys. J. 2003, 84, 4023–4032. [CrossRef]
55. Dickerson, E.B.; Dreaden, E.C.; Huang, X.; El-Sayed, I.H.; Chu, H.; Pushpanketh, S.; McDonald, J.F.;
El-Sayed, M.A. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous
cell carcinoma in mice. Cancer Lett. 2008, 269, 57–66. [CrossRef] [PubMed]
56. Vankayala, R.; Lin, C.C.; Kalluru, P.; Chiang, C.S.; Hwang, K.C. Gold nanoshells-mediated bimodal
photodynamic and photothermal cancer treatment using ultra-low doses of near infra-red light. Biomaterials
2014, 35, 5527–5538. [CrossRef] [PubMed]
57. Cheng, Y.; Samia, A.C.; Meyers, J.D.; Panagopoulos, I.; Fei, B.; Burda, C. Highly efficient drug delivery with
gold nanoparticle vectors for in vivo photodynamic therapy of cancer. J. Am. Chem. Soc. 2008, 130, 10643–10647.
[CrossRef] [PubMed]
58. Lucky, S.S.; Soo, K.C.; Zhang, Y. Nanoparticles in Photodynamic Therapy. Chem. Rev. 2015, 115, 1990–2042.
[CrossRef] [PubMed]
59. Allison, R.; Moghissi, K.; Downie, G.; Dixon, K. Photodynamic therapy (PDT) for lung cancer.
Photodiagn. Photodyn. Ther. 2011, 8, 231–239. [CrossRef] [PubMed]
60. Gondivkar, S.M.; Gadbail, A.R.; Choudhary, M.G.; Vedpathak, P.R.; Likhitkar, M.S. Photodynamic treatment
outcomes of potentially-malignant lesions and malignancies of the head and neck region: A systematic
review. J. Investig. Clin. Dent. 2018, 9, e12270. [CrossRef] [PubMed]
61. Kostovic, K.; Pastar, Z.; Ceovic, R.; Mokos, Z.B.; Buzina, D.S.; Stanimirovic, A. Photodynamic therapy in
dermatology: Current treatments and implications. Coll. Antropol. 2012, 36, 1477–1481. [PubMed]
62. Khlebtsov, B.; Panfilova, E.; Khanadeev, V.; Bibikova, O.; Terentyuk, G.; Ivanov, A.; Rumyantseva, V.;
Shilov, I.; Ryabova, A.; Loshchenov, V.; et al. Nanocomposites containing silica-coated gold-silver
nanocages and Yb-2,4-dimethoxyhematoporphyrin: Multifunctional capability of IR-luminescence detection,
photosensitization, and photothermolysis. ACS Nano 2011, 5, 7077–7089. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2018, 19, 1979 15 of 16
63. Gao, L.; Fei, J.; Zhao, J.; Li, H.; Cui, Y.; Li, J. Hypocrellin-Loaded Gold Nanocages with High Two-Photon
Efficiency for Photothermal/Photodynamic Cancer Therapy in Vitro. ACS Nano 2012, 6, 8030–8040. [CrossRef]
[PubMed]
64. Seo, S.H.; Kim, B.M.; Joe, A.; Han, H.W.; Chen, X.; Cheng, Z.; Jang, E.S. NIR-light-induced surface-enhanced
Raman scattering for detection and photothermal/photodynamic therapy of cancer cells using methylene
blue-embedded gold nanorod@SiO2 nanocomposites. Biomaterials 2014, 35, 3309–3318. [CrossRef] [PubMed]
65. Menon, J.U.; Jadeja, P.; Tambe, P.; Vu, K.; Yuan, B.; Nguyen, K.T. Nanomaterials for photo-based diagnostic
and therapeutic applications. Theranostics 2013, 3, 152–166. [CrossRef] [PubMed]
66. Tomic, S.; Ethokic, J.; Vasilijic, S.; Ogrinc, N.; Rudolf, R.; Pelicon, P.; Vucevic, D.; Milosavljevic, P.; Jankovic, S.;
Anzel, I.; et al. Size-dependent effects of gold nanoparticles uptake on maturation and antitumor functions
of human dendritic cells in vitro. PLoS ONE 2014, 9, e96584. [CrossRef] [PubMed]
67. Qiu, T.A.; Bozich, J.S.; Lohse, S.E.; Vartanian, A.M.; Jacob, L.M.; Meyer, B.M.; Gunsolus, I.L.; Niemuth, N.J.;
Murphy, C.J.; Haynes, C.L.; et al. Gene expression as an indicator of the molecular response and toxicity
in the bacterium Shewanella oneidensis and the water flea Daphnia magna exposed to functionalized gold
nanoparticles. Environ. Sci. Nano 2015, 2, 615–629. [CrossRef]
68. Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-Based Medicines: A Review of
FDA-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016, 33, 2373–2387. [CrossRef] [PubMed]
69. Huhn, D.; Kantner, K.; Geidel, C.; Brandholt, S.; De Cock, I.; Soenen, S.J.; Rivera Gil, P.; Montenegro, J.M.;
Braeckmans, K.; Mullen, K.; et al. Polymer-coated nanoparticles interacting with proteins and cells: Focusing
on the sign of the net charge. ACS Nano 2013, 7, 3253–3263. [CrossRef] [PubMed]
70. Deng, J.; Yao, M.; Gao, C. Cytotoxicity of gold nanoparticles with different structures and surface-anchored
chiral polymers. Acta Biomater. 2017, 53, 610–618. [CrossRef] [PubMed]
71. Libutti, S.K.; Paciotti, G.F.; Byrnes, A.A.; Alexander, H.R., Jr.; Gannon, W.E.; Walker, M.; Seidel, G.D.;
Yuldasheva, N.; Tamarkin, L. Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal
gold-rhTNF nanomedicine. Clin. Cancer Res. 2010, 16, 6139–6149. [CrossRef] [PubMed]
72. Ali, M.R.; Rahman, M.A.; Wu, Y.; Han, T.; Peng, X.; Mackey, M.A.; Wang, D.; Shin, H.J.; Chen, Z.G.; Xiao, H.;
et al. Efficacy, long-term toxicity, and mechanistic studies of gold nanorods photothermal therapy of cancer
in xenograft mice. Proc. Natl. Acad. Sci. USA 2017, 114, E3110–E3118. [CrossRef] [PubMed]
73. Anselmo, A.C.; Samir, M. Nanoparticles in the clinic. Bioeng. Transl. Med. 2016, 1, 10–29. [CrossRef] [PubMed]
74. Kharlamov, A.N.; Tyurnina, A.E.; Veselova, V.S.; Kovtun, O.P.; Shur, V.Y.; Gabinsky, J.L. Silica-gold
nanoparticles for atheroprotective management of plaques: Results of the NANOM-FIM trial. Nanoscale
2015, 7, 8003–8015. [CrossRef] [PubMed]
75. Xu, Z.Q.; Broza, Y.Y.; Ionsecu, R.; Tisch, U.; Ding, L.; Liu, H.; Song, Q.; Pan, Y.Y.; Xiong, F.X.; Gu, K.S.;
et al. A nanomaterial-based breath test for distinguishing gastric cancer from benign gastric conditions.
Br. J. Cancer 2013, 108, 941–950. [CrossRef] [PubMed]
76. Feng, Z.V.; Gunsolus, I.L.; Qiu, T.A.; Hurley, K.R.; Nyberg, L.H.; Frew, H.; Johnson, K.P.; Vartanian, A.M.;
Jacob, L.M.; Lohse, S.E.; et al. Impacts of gold nanoparticle charge and ligand type on surface binding and
toxicity to Gram-negative and Gram-positive bacteria. Chem. Sci. 2015, 6, 5186–5196. [CrossRef] [PubMed]
77. Cho, T.J.; MacCuspie, R.I.; Gigault, J.; Gorham, J.M.; Elliott, J.T.; Hackley, V.A. Highly stable positively
charged dendron-encapsulated gold nanoparticles. Langmuir 2014, 30, 3883–3893. [CrossRef] [PubMed]
78. Schaeublin, N.M.; Braydich-Stolle, L.K.; Schrand, A.M.; Miller, J.M.; Hutchison, J.; Schlager, J.J.; Hussain, S.M.
Surface charge of gold nanoparticles mediates mechanism of toxicity. Nanoscale 2011, 3, 410–420. [CrossRef]
[PubMed]
79. Hanna, S.K.; Montoro Bustos, A.R.; Peterson, A.W.; Reipa, V.; Scanlan, L.D.; Hosbas Coskun, S.; Cho, T.J.;
Johnson, M.E.; Hackley, V.A.; Nelson, B.C.; et al. Agglomeration of Escherichia coli with Positively Charged
Nanoparticles Can Lead to Artifacts in a Standard Caenorhabditis elegans Toxicity Assay. Environ. Sci. Technol.
2018, 52, 5968–5978. [CrossRef] [PubMed]
80. Ginzburg, A.L.; Truong, L.; Tanguay, R.L.; Hutchison, J.E. Synergistic Toxicity Produced by Mixtures of
Biocompatible Gold Nanoparticles and Widely Used Surfactants. ACS Nano 2018, 12, 5312–5322. [CrossRef]
[PubMed]
81. Bahamonde, J.; Brenseke, B.; Chan, M.Y.; Kent, R.D.; Vikesland, P.J.; Prater, M.R. Gold Nanoparticle Toxicity
in Mice and Rats: Species Differences. Toxicol. Pathol. 2018, 46, 192623318770608. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2018, 19, 1979 16 of 16
82. Gao, W.; Xu, K.; Ji, L.; Tang, B. Effect of gold nanoparticles on glutathione depletion-induced hydrogen
peroxide generation and apoptosis in HL7702 cells. Toxicol. Lett. 2011, 205, 86–95. [CrossRef] [PubMed]
83. Rosli, N.S.B.; Rahman, A.A.; Aziz, A.A.; Shamsuddin, S. Determining the size and concentration dependence
of gold nanoparticles in vitro cytotoxicity (IC50 ) test using WST-1 assay. AIP Conf. Proc. 2015, 1657. [CrossRef]
84. Connor, E.E.; Mwamuka, J.; Gole, A.; Murphy, C.J.; Wyatt, M.D. Gold nanoparticles are taken up by human
cells but do not cause acute cytotoxicity. Small 2005, 1, 325–327. [CrossRef] [PubMed]
85. Zhang, X.D.; Wu, D.; Shen, X.; Chen, J.; Sun, Y.M.; Liu, P.X.; Liang, X.J. Size-dependent radiosensitization of
PEG-coated gold nanoparticles for cancer radiation therapy. Biomaterials 2012, 33, 6408–6419. [CrossRef]
[PubMed]
86. Li, X.; Hu, Z.; Ma, J.; Wang, X.; Zhang, Y.; Wang, W.; Yuan, Z. The systematic evaluation of size-dependent
toxicity and multi-time biodistribution of gold nanoparticles. Colloids Surf. B Biointerfaces 2018, 167, 260–266.
[CrossRef] [PubMed]
87. Sonavane, G.; Tomoda, K.; Makino, K. Biodistribution of colloidal gold nanoparticles after intravenous
administration: Effect of particle size. Colloids Surf. B Biointerfaces 2008, 66, 274–280. [CrossRef] [PubMed]
88. Fraga, S.; Brandao, A.; Soares, M.E.; Morais, T.; Duarte, J.A.; Pereira, L.; Soares, L.; Neves, C.; Pereira, E.;
Bastos Mde, L.; et al. Short- and long-term distribution and toxicity of gold nanoparticles in the rat after
a single-dose intravenous administration. Nanomedicine 2014, 10, 1757–1766. [CrossRef] [PubMed]
89. Cho, W.S.; Cho, M.; Jeong, J.; Choi, M.; Cho, H.Y.; Han, B.S.; Kim, S.H.; Kim, H.O.; Lim, Y.T.; Chung, B.H.; et al.
Acute toxicity and pharmacokinetics of 13 nm-sized PEG-coated gold nanoparticles. Toxicol. Appl. Pharmacol.
2009, 236, 16–24. [CrossRef] [PubMed]
90. Zharov, V.P.; Kim, J.W.; Curiel, D.T.; Everts, M. Self-assembling nanoclusters in living systems: Application
for integrated photothermal nanodiagnostics and nanotherapy. Nanomedicine 2005, 1, 326–345. [CrossRef]
[PubMed]
91. Cheng, Z.; Al Zaki, A.; Hui, J.Z.; Muzykantov, V.R.; Tsourkas, A. Multifunctional nanoparticles: Cost versus
benefit of adding targeting and imaging capabilities. Science 2012, 338, 903–910. [CrossRef] [PubMed]
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).