Вы находитесь на странице: 1из 10

Review

Near-infrared fluorescent nanoprobes


for cancer molecular imaging:
status and challenges
Xiaoxiao He1,2*, Jinhao Gao1,3*, Sanjiv Sam Gambhir1,4 and Zhen Cheng1
1
Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Bio-X Program and Stanford Cancer Center,
Stanford University School of Medicine, Stanford, CA 94305, USA
2
State Key Laboratory of Chemo/Biosensing and Chemometrics, Institute of Biology, Hunan University, Changsha 410082,
P.R. China
3
Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P.R. China
4
Departments of Radiology and Bioengineering, Stanford University, Stanford, CA 94305, USA

Near-infrared fluorescence (NIRF) imaging promises to low absorption and relatively low autofluorescence, it offers
improve cancer imaging and management; advances in several advantages over other modalities for imaging living
nanomaterials allow scientists to combine new nano- organisms. In addition, NIRF imaging has potentially high
particles with NIRF imaging techniques, thereby fulfill- spatial resolution, high sensitivity and low risk to the living
ing this promise. Here, we present a synopsis of current subject because it utilizes nonionizing radiation. Moreover,
developments in NIRF nanoprobes, their use in imaging it is cost-efficient in terms of preparation of molecular probes
small living subjects, their pharmacokinetics and toxici- and the detection hardware is relatively simple to operate.
ty, and finally their integration into multimodal imaging The integration of nanotechnology with molecular biolo-
strategies. We also discuss challenges impeding the gy and medicine has resulted in active developments of an
clinical translation of NIRF nanoprobes for molecular emerging research area, namely nanobiotechnology. This
imaging of cancer. Whereas utilization of most NIRF research offers exciting and abundant opportunities for
nanoprobes remains at a proof-of-principle stage, opti- discovering new materials and tools for biomedicine. Recent
mizing the impact of nanomedicine in cancer patient advancements in functional nanomaterials offer to improve
diagnosis and management will probably be realized detection sensitivity and specificity in molecular imaging
through persistent interdisciplinary amalgamation of [14]. Functional nanomaterial-based molecular probes,
diverse research fields. namely nanoprobes, could target tumors either through
the enhanced permeability and retention (EPR) effect of
Molecular imaging and nanotechnology the tumor microvasculature or by the specific binding with
Cancer molecular imaging is an evolving field in which tumor-associated biomarkers, such as tumor cell receptors,
diverse optical tools and strategies are used for early tumor extracellular matrix and enzymes. A variety of
detection and management of tumors. This field arose from nanoprobes have been prepared, evaluated and applied in
the merger of several pre-existing disciplines, such as various imaging modalities, including fluorescence, MRI,
modern cancer molecular biology, chemistry and imaging radionuclide, Raman and photoacoustic imaging. Examples
technologies. Consequently, cancer molecular imaging has of probes include gold (Au) nanoparticles (nanoshells, nanor-
created unique opportunities to study and noninvasively ods and nanocages) or single wall carbon nanotubes
monitor tumor genesis, development and metastasis in (SWNTs) for photoacoustic imaging based on heating effects
vivo [1,2]. It is expected to provide more comprehensive [15,16]; magnetic iron oxide nanoparticles for MRI [17]; and
anatomical, physiological and functional information of fluorescent nanoparticles for in vivo fluorescence imaging
diseases in a clinical setting. Molecular imaging techni- [18]. Nanoparticle-based NIRF probes can overcome several
ques could be powerful tools in early cancer detection, drug limitations of conventional NIR organic dyes, such as poor
discovery and development, as well as monitoring response hydrophilicity and photostability, low quantum yield and
to treatment [3–5]. There are a variety of well-established
imaging modalities, such as positron emission tomography
(PET) [3,6], single photon emission computed tomography Glossary
(SPECT) [7], magnetic resonance imaging (MRI) [8–10] Fermi wavelength: the size scale is related to EFermi/N1/3, predicted by the free-
and optical fluorescence imaging [11–13], that effectively electron model of metallic behavior (approximately 0.5 nm for gold and silver).
EFermi (Fermi energy) is the energy of the highest occupied quantum state in a
image specific tumor-associated molecular targets. system of fermions at absolute zero temperature.
Here, we describe the development of near-infrared fluo- Fluorescence-mediated tomography (FMT): a method of molecular imaging
rescence (NIRF) nanoprobes for cancer molecular imaging. shows the distribution of a NIRF probes in the region of an animal by three-
dimensional tomographic images.
Because NIRF imaging (650–900 nm) displays properties of Raman signatures: the Raman spectrum of a particular material shows the
‘‘fingerprints’’, which are known as spectra signatures and enable identifica-
Corresponding author: Cheng, Z. (zcheng@stanford.edu) tion of the materials that make up a scanned object.
*
These authors contributed equally.

574 1471-4914/$ – see front matter ß 2010 Elsevier Ltd. All rights reserved. doi:10.1016/j.molmed.2010.08.006 Trends in Molecular Medicine, December 2010, Vol. 16, No. 12
(Figure_1)TD$IG][ Review Trends in Molecular Medicine Vol.16 No.12

somes, micelles and dendrimers, have been used for drug


delivery research for years. Organic systems that encapsu-
late NIRF dyes for cancer molecular imaging have also
been reported [22–24], for example the water-soluble poly-
meric nanoparticles were used as carriers of NIRF dyes for
tumor imaging [25,26].
Compared to the bare NIR fluorochromes, there are
several distinctive features in the NIRF dye-containing
nanoprobes. Firstly, the polymer or inorganic matrix en-
capsulation provides a protective layer around NIRF
molecules that reduces their exposure to oxygen both in
air and in aqueous media. Encapsulation also prevents
direct interaction between NIRF dyes and host tissue. As a
result, photostability and biocompatibility of NIRF dye-
containing nanoprobes in vivo increase substantially in
comparison to the bare dyes. Secondly, by using well-estab-
Figure 1. Representative nanomaterial-based NIRF nanoprobes for molecular lished bioconjugation chemistry, the surface of dye-doped
imaging in living subjects. nanoparticles can be easily modified with a variety of
biomolecules, such as proteins, peptides, antibodies or oli-
detection sensitivity, insufficient stability in biological sys- gonucleotides. These biomolecules affect the pharmacoki-
tems and weak multiplexing capability. netics of the nanoparticle and make specific molecular
In this short review, we focus on the latest progress in targeting possible. Thirdly, the NIRF signal can be en-
NIRF nanoprobes for cancer molecular imaging. More hanced because of the high payload of dye molecules per
specifically, we summarize the development of different nanoparticle, which results in much higher sensitivity com-
types of NIRF nanoprobes, their use for molecular imaging pared to free fluorochromes [25]. For example, approximate-
in living subjects, the potential toxicity of NIRF nanop- ly 600 indocyanine green (ICG) molecules can be doped into
robes and the combination of NIRF nanoprobe imaging each CPNP, exhibiting significantly greater intensity at the
with several other imaging techniques. We also discuss maximum emission wavelength relative to the free constit-
future perspectives of the NIRF nanoprobes for cancer uent fluorophore [20].
molecular imaging.
NIR fluorescent and self-illuminating QDs
Development of NIRF nanoprobes QDs are a special class of materials known as semiconduc-
Although the number of NIRF nanoprobes is rapidly in- tors. Because of their unique optical properties, such as
creasing, most of them can be classified into two major size-tunable photoluminescent emission, narrow and sym-
categories: downconversion (DCN) and upconversion metric emission spectra, broad absorption spectra and
(UCN) NIRF nanoprobes. DCN nanoprobes produce low photostability, the use of QDs as fluorescent biotags for
energy fluorescence when they are excited by high energy cell staining and disease detection is one of the most
light. The well-established DCN NIRF nanoprobes include attractive applications in nanomedicine research [27–
NIRF dye-containing nanoparticles, quantum dots (QDs), 29]. The synthesis, surface modification, characterization
SWNTs and metal nanoclusters (Figure 1). By contrast, and application of QDs in the visible spectrum have been
UCN nanoprobes, which are emerging as a new class of reviewed previously [30–32]. As a new class of fluorescent
fluorescent nanoparticles and biolabels [19], can convert probes, QDs could overcome the limitations of organic dyes
excitation light with longer wavelength (low energy) to and they have high potential for cancer molecular imaging.
shorter wavelength fluorescence (high energy). In this Still, the main barriers preventing the in vivo application
section, we focus on well-established NIRF nanoprobes: of QDs excited by an external illumination source in the
NIRF dye-containing nanoparticles, NIR fluorescent QDs, visible spectrum are autofluorescence and light scattering
SWNTs and Au nanoclusters. within tissues. Therefore, NIRF-emitting QDs have been
the focus of increased attention because, with NIRF QDs,
NIRF dye-containing nanoprobes tissue autofluorescence is considerably reduced and the
NIRF dye-containing nanoprobes are organic or inorganic tissue penetration of the excitation light is significantly
matrix-based nanomaterials that either incorporate NIRF enhanced [33]. Because of high interest and demand for
dyes inside the matrix (dye-doped) or attach the reporting NIRF-emitting QDs, the development and the synthesis of
molecules to the nanoparticles surface [18]. The matrix NIR QDs, including cadmium (Cd) and noncadmium based
should be optically transparent to allow excitation and QDs, have progressed rapidly. Examples include CdTe
emission light to pass efficiently. Inorganic matrices for (cadmium telluride)/ZnS (zinc sulfide) [34], CdTe/CdSe
dye encapsulation encompass mainly silica and calcium (cadmium selenide) [35], InAs (indium arsenide)/InP (indi-
phosphate nanoparticles (CPNPs) [20,21]. Silica matrix is um phosphide)/ZnSe [36], CuInSe2 (copper indium disele-
attractive for in vivo imaging because its hydrophilic nide) [37] and Cu-doped InP/ZnSe [38] QDs.
nature can reduce nonspecific binding and aggregation; However, Cd-based QDs showed cytotoxicity in vitro
it is also chemically inert, transparent and easily modified under extreme conditions [39,40]. To avoid such toxicity
through chemistry. Organic nanocarriers, such as lipo- problems, the Cd metal can be replaced by other more

575
Review Trends in Molecular Medicine Vol.16 No.12

benign elements. For example, CuInS2/ZnS core/shell QDs another study, radiation produced light with broad contin-
do not contain any class A elements [e.g. Cd, Pb (lead) and uous wavelengths (ultraviolet-visible to NIR), which can
Hg (mercury)] [41] and have potential as biocompatible excite fluorescent materials such as QDs (Figure 2c). This
probes for biomedical applications. Recently, novel optical provides an alternative strategy for the design of self-
probes such as carbon dots and silicon nanoparticles have illuminating nanoprobes. By marrying this technique with
aroused interest because of their low toxicity [42–44]. the intrinsic property of QDs (e.g. the capacity of simulta-
In addition to the tunability of emission in the NIR neous excitation of multiple fluorescent colors), multiplex-
region, the amount of incident excitation light that reaches ing in vivo imaging with radionuclides (e.g. 131Iodine) can
fluorescent objects in vivo can be limited. Bioluminescence become a reality (Figure 2d). Radiation-luminescence
resonance energy transfer (BRET), which converts chemi- excited nanoprobes might have advantages in multiplex
cal energy from a chemical reaction catalyzed by a donor fluorescence imaging as well as multimodality imaging
enzyme rather than the absorption of excitation photons [51].
into light energy, has been used to circumvent this issue
[45,46]. So et al. reported self-illuminating NIR QDs by Carbon nanotubes
BRET without the need of external illumination [47–49]. In Carbon nanotubes (CNTs) can be broadly classified as
these studies, QDs, the acceptor in a BRET system, were SWNTs and multiwalled nanotubes (MWNTs). Both cate-
covalently conjugated to the donor, Renilla luciferase gories have attracted interest for their unique electronic,
(Luc8) [50]. Upon interaction with its substrate coelenter- metallic and structural characteristics. Traditionally, most
azine, the Luc8 protein emits blue light with a peak at research on CNTs has focused on their mechanical, ther-
480 nm. This light excites QD Luc8 conjugates to produce mal and electronic properties and their potential applica-
NIR light (Figure 2a). The key advantage of using biolu- tion in electronic devices. Since 2002 and the discovery of
minescence is the elimination of autofluorescence as no fluorescent semiconducting CNTs, research into CNTs for
external light excitation is needed (Figure 2a,b). This ‘‘self- their optical properties and their application to bioimaging
illuminating’’ feature could allow molecular imaging of have accelerated [52–55].
cancer in deeper tissue. Because no excitation light is SWNTs can be viewed as a rolled graphene (a single
needed, light scattering from the tissue and the toxicity plane of graphite sheet) and are classified by a vector
to the tissue from excitation photons are also avoided. connecting the two points that meet upon rolling
However, substrate must still be delivered for BRET. In (Figure 1). The rolled single tube is approximately 1 nm
[(Figure_2)TD$IG]

Figure 2. Self-illuminating nanoprobes based on NIRF QDs. (a) Molecular imaging of bioluminescent QD conjugates in small animals based on BRET. The scheme shows a
QD that is covalently coupled to a BRET donor, Luc8. (b) Representative bioluminescence images of a nude mouse injected via tail vein with labeled cells, acquired with a
filter (575–650 nm) (left) and without any filter (right). Reprinted with permission from [47]. Copyright 2006, Nature Publishing Group. (c) The scheme shows the radiation
luminescence excited QDs for optical imaging. (d) Multiplexed in vivo radioactivity illuminated QD imaging shows the spectral imaging of the QD655, QD705 and QD800 in
mice, respectively, and the spectral unmixed image of the same mouse (right). Reprinted with permission from [51]. Copyright 2010, Wiley-VCH.

576
Review Trends in Molecular Medicine Vol.16 No.12

in diameter and several hundred nanometers to 100 mm in have been applied for tumor targeting of NIRF nanopar-
length. Depending on how a nanotube is wrapped up from a ticles: (i) the well-known EPR effect; (ii) molecular target-
single plane of graphene, it can be semiconducting or ing via specific antigens or overexpressed receptors on the
metallic. SWNTs are promising NIRF nanomaterials for surface of cancer cells; and (iii) the chemical activation of
molecular imaging because they have the following fea- the nanoprobes in specific tumor microenvironments (e.g.
tures. First, the emission of SWNT-based NIRF probes are enzymatic cleavage or oxidation).
mostly in the infrared-A region (1–1.4 mm), making them
ideal for in vivo imaging because of low biological auto- Passive targeting of NIRF nanoprobes
fluorescence. Second, there is a large Stoke’s shift between Compared with normal cells, cancer cells tend to proliferate
the excitation and emission bands of SWNTs, making it rapidly and secrete factors that promote the formation of
possible to excite the nanoparticles in the NIR window tumor-associated neovasculature from the existing vascula-
thereby reducing further autofluorescence and scattering. ture. This produces the well-known EPR effect; the tumor-
Third, SWNTs possess high surface area and can load large associated neovasculature has large interendothelial junc-
amounts of targeting ligands or drugs. Finally, SWNTs are tions, an imperfect basement membrane, an inefficient
highly photostable, unlike conventional fluorophores, lymphatic system and large numbers of transendothelial
which allows them to serve as long-term NIRF tracking channels. Furthermore, tumor tissues tend to have poor
probes in vivo [54]. However, the fluorescence of SWNTs lymphatic drainage. Selective targeting of tumor tissue with
can be quenched when they are aggregated in bundles. nanoparticles can be achieved by taking advantage of the
Biocompatible SWNTs with high quantum yield are elu- EPR effect of the tumor microvasculature and the poor
sive, which might limit their use in cancer fluorescence lymphatic drainage of tumors [65]. Nanoparticles ranging
imaging. In the future, chemical modification will play a from 10 to 100 nm might escape renal filtration and accu-
key role in the fabrication of high performance of SWNTs mulate in tumors after prolonged circulation [66]. The level
for optical imaging. of nanoparticle accumulation in tumors depends both on the
properties of the nanoparticles and the type of the tumor.
Au nanoclusters Variables include the size, surface modification and the half-
Nanometer-sized Au particles are one of the most impor- life of the nanoparticles in circulation, as well as the leaki-
tant classes of metal nanoparticles and have been used ness of vascular pore, the degree of tumor vascularization
widely to stain cells and tissue samples for electron mi- and the degree of angiogenesis in the tumor. NIRF dye-
croscopy. Au nanoparticles do not emit fluorescence but containing nanoparticles have much longer circulation
effectively scatter light, exhibiting a range of intense colors times than free NIRF dyes, which by consequence
in the visible and NIR spectra. The Au nanoparticle-based encourages the accumulation in tumors through the EPR
bioanalytic method is a promising technique for detecting a effect. This accumulation enhances the imaging signal,
wide range of biological macromolecules because it offers which comes from both the selective trapping of the nano-
distinct Raman signatures (see Glossary) and easy surface particles as well as an amplification and protection effect
functionalization. Au-based nanomaterials, such as nano- owing to their encapsulation. Altinoglu et al. [20] used
particles, nanoshells and nanorods, have also been used as biodegradable CPNPs encapsulated with the NIR-emitting
contrast agents for imaging cancer in vivo by taking fluorophore ICG as a new fluorescent nanoprobe for sensi-
advantage of their surface plasmon bands in the NIR tive diagnostic imaging. The CPNPs with an average size of
region [56–59]. Interestingly, emission from small Au 16 nm in diameter prolonged circulation time in vivo with
nanoclusters with sizes below 2 nm has gained significant high passive tumor accumulation of the nanoparticles in
attention. All Au nanoclusters are a collection of a small solid xenograft breast adenocarcinoma tumors within 24 h
number of Au atoms (Figure 1), which have sizes compa- of systemic tail vein injection [20].
rable to the Fermi wavelength (see Glossary), resulting in For the passive delivery of QDs, the efficiency of target-
molecule-like behavior including discrete electronic states ing relies on the inherent physicochemical properties of the
and size-dependent fluorescence [60]. Au nanoclusters are QDs (e.g. particle size, charge and surface properties). One
attractive for bioimaging applications because of their critical issue for applications of QDs in living subjects is the
potential for low toxicity and their ultrafine size. Au hydrodynamic diameter (HD) of nanoparticles. QDs with
nanoclusters with emissions from blue to red have been large diameters (>20 nm) endure extremely high uptake in
reported by using poly(amidoamine) dendrimer, mercap- both the reticuloendothelial system (RES) and the mono-
toundecanoic acid and lipoic acid as nanoparticle tem- nuclear phagocytic system (MPS), which reduces their
plates or ligands [61–63]. Recently, advances in green efficiency and sensitivity. Ultrasmall water-soluble QDs
chemistry with environmentally friendly methods have (<10 nm) have been the focus of increased attention be-
made it possible to prepare NIR Au nanoclusters using cause their unique properties, rapid renal clearance
bovine serum albumin (BSA) as a template at physiological [43,44,67,68], low RES uptake and the possibility of EPR
temperature [64]. The emergence of NIRF Au nanoclusters effect, are advantageous for in vivo applications. Recently,
that possess advantages of both small size and brightness Gao et al. reported high tumor uptake of ultrasmall NIR
opens a new path toward in vivo imaging applications. noncadmium QDs owing to the EPR effect [69].
Because of the small size of vessels and poor access, in
NIRF nanoprobes for cancer molecular imaging vivo visualization of the lymphatic function is relatively
One of the major concerns for imaging with NIRF nano- difficult. Kim et al. reported the utility of NIR QDs (CdTe/
particles in living subjects is specificity. Three approaches CdSe) for sentinel lymph node imaging in both mouse and

577
Review Trends in Molecular Medicine Vol.16 No.12

porcine models. QDs translocated to sentinel lymph nodes, of NIRF nanoprobes, such as small molecules, peptides,
probably from the combination of passive flow in lymphatic proteins, aptamers, engineered antibodies and antibody-
vessels and the active migration of dendritic cells that based ligands [12,13,29]. Targeted NIRF nanoprobes
engulfed the nanoparticles [35]. Subsequently, Hama might significantly improve the efficiency (specificity and
et al. showed simultaneous in vivo two-color wave- selectivity) of delivery to the tissue of interest.
length-resolved spectral fluorescence lymphangiography Using small animals as models, Gao et al. first described
using two NIR CdTe-based QDs with different emission in vivo cancer targeting with QD antibody conjugates [72].
spectra (705 and 800 nm) [70]. Two-color spectral fluores- Subsequently, numerous publications have reported tar-
cence lymphangiography can provide insight into drainage geted molecular imaging of cancer via QDs with peptides
from different lymphatic basins that can detect sentinel and proteins as the main targeting biomolecules. In vivo
lymph nodes of breast cancer and prevent complications targeted imaging of tumor vasculature was achieved using
such as lymphedema of the arm [70]. peptide-conjugated NIR QDs [73–75] through surface mod-
Although SWNTs are relatively long (0.5–2 mm), in vivo ification of NIR QDs with arginine–glycine–aspartic acid
fluorescence imaging by semiconducting SWNTs is still (RGD) peptides to produce constructs known as QD–RGDs.
possible owing to their flexible structure. Welsher et al. The QD–RGDs showed specific and strong binding to
developed bright fluorescent SWNTs by sonicating SWNTs integrin avb3-positive vasculature to human xenografts
with sodium cholate, followed by surfactant exchange to implanted into nude mice. However, the relatively large
form phospholipid–polyethylene glycol (PEG) coated nano- size of the QD–RGD (20 nm in HD) prevented efficient
tubes. Using this type of SWNT, high-resolution intravital extravasation, thus QD–RGD mainly targeted the tumor
images of tumor vessels below the skin of mice were vasculature instead of the tumor cells. The in vivo target-
obtained [71]; these SWNTs might have applications in ing behavior of QD–RGD has also been demonstrated in
image-guided surgery. tumor vessels by intravital imaging of QD800–RGD in real
time [76]. As shown in Figure 3, QD800–RGD does not
Targeted molecular imaging of NIRF nanoprobes extravasate in an SKOV-3 mouse ear tumor model;
In contrast to gross structural imaging, the advantage of QD800–RGDs specifically bind the tumor neovasculature
molecular imaging is the potential to reveal differences as aggregates, but no binding occurs for the QD800 modi-
between cancerous and normal tissues at the molecular fied with a control peptide RAD. However, the behavior of
level. To realize this potential, NIRF nanoprobes need to be QDs and other nanoparticles can vary significantly be-
modified with biomolecules that facilitate binding only in tween different tumor models. The high reproducibility
the targeted locations. A variety of targeting groups have of QDs bioconjugated with RGD peptides and the feasibili-
been developed for the modification and functionalization ty of QD–RGDs as tumor-targeted fluorescence nanoprobes
[(Figure_3)TD$IG]

Figure 3. Direct visualization of QD800–RGD binding to tumor vessel endothelium and controls in a living mouse ear tumor model using intravital microscopy. (a) Each
panel displays different output channels of the identical imaging plane along the row. In the green channel, individual enhanced green fluorescent protein-expressing
cancer cells are visible, whereas the red channel shows the vasculature of the tumor via angiosense dye injection. The NIR channel shows intravascularly administered QDs
that remain in the vessels. Binding events are visible by a bright white signal. These are demarcated by arrows in the rightmost merged image in which all three channels
have been overlaid. (b) Merged image of a different mouse using QD800–RGD. Individual cells are not generally visible. (c–e) Typical images without binding in each control
condition: (c) tumor neovasculature containing unconjugated QDs, (d) normal vasculature containing QD800–RGD and (e) tumor neovasculature containing QD800–RAD.
Reprinted with permission from [76]. Copyright 2008, American Chemical Society.

578
Review Trends in Molecular Medicine Vol.16 No.12

warrant the applications of QDs to molecular imaging and distinct properties makes it possible to accomplish multi-
diagnosis in living subjects. modal imaging [83–85]. The future of cancer molecular
imaging will increasingly rely upon multimodality and
Activatable NIRF nanoprobes nanoplatform-based imaging will be one of the most im-
Interdisciplinary research between scientists in the fields portant approaches in multimodality imaging. The combi-
of molecular imaging and bionanoconjugation chemistry nation of NIRF imaging with nuclear imaging techniques,
has generated activatable imaging nanoprobes with high- such as PET and SPECT, or anatomical imaging modali-
resolution imaging capabilities and ultralow background ties, such as MRI and CT (computed tomography), could
signals. Activatable imaging probes amplify output signals provide more accurate physiological and spatial informa-
in response to a specific biomolecular recognition or an tion, which is imperative for early cancer detection and
environmental change in real time [77]. The unique char- cancer management.
acteristics of activatable fluorescence molecular probes The combination of NIRF nanoprobes with MRI is a
could provide higher signal-to-background ratios com- major research effort. Examples include the combination of
pared to conventional ‘‘constantly active’’ fluorescent NIRF nanoprobes with superparamagnetic iron oxide or
probes. Nanoparticle-based activatable probes have gadolinium MR contrast materials [86–88]. In addition to
attracted significant attention because they could offer MRI, radionuclide imaging can also be combined with
considerable advantages for obtaining optical images NIRF imaging, allowing increased depth penetration
through their NIRF quenching properties. The design of and absolute quantification. Recently, Cai et al. and Chen
activatable nanoprobes relies on specific tumor-related et al. described the quantitative tumor targeting efficacy of
enzymes, such as cathepsin D and matrix metalloprotei- dual-functional QD-based probes using both PET and
nases-2 (MMP-2) [78–80]. NIRF imaging [89,90]. Dual-modality PET/NIRF imaging
There are mainly two types of nanoparticle-based acti- probes offer synergistic advantages over the single modal-
vatable probes. One involves energy transfer between ity probes by overcoming the low quantitative analysis of
NIRF dyes and nanoparticles with quenching properties. fluorescence intensity in vivo and ex vivo. Nahrendorf et al.
The polymer-based nanoparticles and Au nanoparticles demonstrated the exquisite congruence between optical
are the most widely applied quenching moieties [81]. For fluorescence-mediated tomography (FMT; see Glossary)
example, Lee et al. engineered 20-nm Au nanoparticles and PET measurements using targeted multimodal nanop-
stabilized by a Cy5.5-labeled peptide substrate, Gly–Pro– robes [91]. Using biocompatible nanoparticles as a generic
Leu–Gly–Val–Arg–Gly–Cys, where the core of the peptide platform, the combination of radionuclides, NIR fluoro-
substrate is selectively cleaved by MMP-2. The stabilized chromes and targeting ligands result in multimodal nanop-
nanoprobe has strong quenching properties with minimal robes (Figure 4). A high correlation can be seen between
background signals. Only in the presence of MMPs in vitro FMT and PET in nanoprobe concentration and spatial
or in vivo when the quenching nanoparticle and the signal distribution both in vitro and in living subjects
fluorescent dye are separated by cleavage of the peptide (Figure 4), which justifies the development of next gener-
do the NIRF nanoprobes recover fluorescence [80]. ation of PET/optical molecular imaging nanoprobes.
The other type of nanoparticle-based activatable probes Moreover, noninvasive PET imaging using radiolabeled
involve energy transfer between QDs and Au nanoparti- nanoprobes can provide a robust and reliable measure of
cles, where QDs serve as donors and Au nanoparticles act the biodistribution of nanoprobes in living subjects [92,93].
as quenchers. The luminescence of the QDs could be re- Although the stability and physicochemical properties of
duced by more than 70% when they were attached to the nanoprobes after radionuclide labeling should be carefully
Au nanoparticles [80]. A conjugated QD–peptide–Au com- examined, the accurate evaluation of in vivo distribution
plex was prepared using a peptide that is a substrate for and tumor targeting efficacy using these dual-modality
collagenase. Upon exposure to collagenase, the peptide is probes could significantly facilitate applications of NIRF
digested, releasing the QDs from the Au complex. The nanoprobes in biomedical research and in the clinic.
quenched (dark) nanoprobe complex is activated and the
luminescence of the QDs is recovered [82]. Although many Pharmacokinetics and toxicity of NIRF nanoprobes
hurdles need to be overcome before QD–peptide–Au con- Comprehensive insights on how NIRF nanoprobes enter,
jugates can be applied as activatable NIRF nanoprobes in distribute and leave living subjects are vital towards de-
living subjects, the potential advantages of this smart class signing NIRF nanoprobes suitable for molecular imaging.
of NIRF nanoprobes should inspire more research in the To have a critical level of NIRF nanoprobes entering the
NIRF field. tumor site, they must avoid uptake by both RES and MPS;
however, many systemically injected NIRF nanoprobes
Multimodality imaging can be rapidly cleared from the bloodstream by RES and
The combination of multiple imaging modalities can yield MPS uptake, leading to accumulation and retention in the
complementary information and offers synergistic advan- liver and spleen. Therefore, the development of NIRF
tages over any single modality. Compared to other imaging nanoprobes that evade rapid blood clearance is a key step
agents, nanoparticles have the advantages of multifunc- in improving their performance as imaging agents. Indeed,
tionality and enormous flexibility, allowing for the integra- surface coating, size and surface charge are three key
tion of multimodality reporting moieties, targeting ligands features that influence the behavior of NIRF nanoprobes
and even therapeutic components into one entity. in living subjects and could guide the design of future NIRF
The development of multifunctional nanomaterials with nanoprobes for cancer molecular imaging.

579
(Figure_4)TD$IG][ Review Trends in Molecular Medicine Vol.16 No.12

Figure 4. Hybrid FMT/PET-CT imaging using multimodal NIRF nanoprobes. (a) The scheme shows nanoplatform-based (dextran-coated iron oxide nanoparticles, CLIO)
nanoprobes containing radionuclides, NIR fluorochromes (e.g. VT680) and affinity ligands, such as peptides and small molecules. (b) The three-dimensional reconstruction
of CT, FMT and PET dataset after injection of 18Fluorine–CLIO–VT680 into a control mouse. Signals are predominantly seen in the liver and spleen. (c) Comparison of FMT-
CT, PET-CT and FMT/PET-CT reconstructions in a representative mouse with bilateral flank tumors showing correlation of spatial distribution and the amplitude of signals.
Reprinted with permission from [91]. Copyright 2010, the National Academy of Sciences.

Surface modification of NIRF nanoprobes with hydro- with nanoprobe behavior in living subjects. Significant
philic polymers such as PEG can enhance solubility, mini- efforts are underway to investigate and optimize these
mize nonspecific binding, prolong circulation time and parameters for improved imaging efficacy [95].
enhance tumor specific targeting [94], but the size of the Understanding and solving the potential in vivo toxicity
resulting nanoparticles can increase significantly. Most of of nanoprobes is critical for their eventual clinical use [96].
the NIRF nanoprobes that have been used for imaging in Toxicity could result from NIRF nanoprobes themselves or
vivo are relatively large (>20 nm in HD) and typically the individual components of the nanoprobes that are
accumulate in the liver. Smaller nanoprobes can be cleared released during degradation in vivo. To mitigate the
by the renal system, which is preferred clinically because potential toxicity, several strategies have been applied
the nanoprobes will be less likely to be sequestered in the to construct biocompatible and biodegradable nanoprobes
body for an extended period of time. QDs with neutral composed of low toxicity building blocks. CPNPs encapsu-
charge and a HD of approximately 5.5 nm might be cleared lated clinically approved ICG fluorophores are biocompati-
completely by the kidneys in a reasonable amount of time ble and biodegradable [20]. Biodegradable NIR silicon
[67]. Although small nanoprobes might display short half- nanoparticles with no detectable toxicity to small animals
lives, this usually results in low accumulation in tumors have also been reported [43]. Dye-containing water-soluble
[12]. NIRF nanoprobes of reasonable size should exhibit polymers might have a chance for clinical use because the
two features: the ability to extravasate and a long enough polymers can be designed to be biocompatible and degrad-
half-life to accommodate extravasation. Nonspecific bind- able [25,26].
ing to serum proteins in the blood, which can be influenced Although Cd-based QDs showed cytotoxicity under ex-
by the surface coating and surface charge of nanoprobes, treme conditions, most toxicology data are derived from in
often results in high RES uptake of nanoparticles. Thus, vitro studies and might not reflect in vivo responses. In vivo
the three characteristics of NIRF nanoparticles size, sur- responses are the ultimate concern for the clinical transla-
face coating and surface charge can mutually interfere tion of nanoprobes. Aillon et al. [95] described systematic

580
Review Trends in Molecular Medicine Vol.16 No.12

toxicity of Cd-based QDs using healthy rats as a model. The coating nanomaterials that can help NIRF nanoprobes
QD formulations (CdSe/ZnS core/shell QDs with polymer, escape from the RES and also provide active functional
PEG or BSA coating) do not cause appreciable toxicity even groups for controllable bioconjugation of specific targeting
after they breakdown in rats over time [97]. Similarly, molecules. For NIRF imaging of deep tissues, further
systematic evaluation of SWNT toxicity in animals revealed innovations in probe preparation are necessary to generate
no evidence of toxicity for over four months following intra- NIRF nanoprobes with high quantum yield and deep tissue
venous injection [98]. Although these data are preliminary penetration. In the clinical setting, optical imaging is
and need confirmation, in aggregate they are encouraging relevant for tissues close to the surface of skin, tissues
and thereby compel us to research further the use of these accessible by endoscopy and intraoperative visualization.
nanoparticles for nanoprobe development. The use of multifunctional nanoparticles (e.g. paramagnet-
ic NIRF nanoprobes) would have great advantages for
Challenges and perspectives providing tumor assessment and intraoperative surgical
Recent and rapid development of synthesis technologies for guidance for tumor tissue resection [101,102], because the
nanomaterials has created enormous opportunities for the combination of NIRF imaging with MRI, PET and X-ray
design of specific and sensitive NIRF nanoprobes for cancer are expected to provide anatomical and biological informa-
molecular imaging. The potential to diagnose and monitor tion with accurate location and quantification of the fluo-
altered physiological changes of cancer in patients by using rescence signal.
NIRF nanoprobes is coming closer to reality. However, In summary, the successful development of NIRF
there remain considerable challenges pertaining to appli- nanoprobes and their potential applications in cancer mo-
cations of NIRF nanoprobes in humans. In addition to the lecular imaging have inspired increasing research inter-
purity, dispensability and stability of NIRF nanoprobes in ests in nanomedicine and nanobiotechnology. The
physiological environments, the variable physicochemical applications of NIRF nanoprobes in medical research have
properties of different NIRF nanoprobes might produce already been significant. Although the practical clinical
unexpected outcomes in vivo. Several studies have shown application of NIRF nanoprobes has proved challenging,
that NIRF nanoprobes might be systemically distributed continued collaborations among the fields of materials
in organs and tissues. Absorption, distribution, metabo- science, chemistry, molecular biology and imaging, preclin-
lism and excretion characteristics are highly variable for ical and clinical medicine and the regulatory authorities
NIRF nanoprobes because of the wide variation in the will no doubt speed up the translational process and thus
physicochemical properties of nanomaterials [95,99,100]. eventually the impact of nanomedicine in the diagnosis
Progress toward the clinical adaptation of NIRF and management of cancer can be realized.
nanoprobe-based molecular imaging might be slow as
probes for clinical use generally undergo stringent itera- Acknowledgments
tive redesign, optimization and validation that can take This research was partially supported by National Cancer Institute/
National Institutes of Health (NCI/NIH) R21 CA121842 (to Z.C.), NCI In
many years. Many issues need to be overcome before the
vivo Cellular and Molecular Imaging Centers (ICMIC) P50 (to S.S.G.) and
NIRF nanoprobes will be ready for clinical translation NCI Center for Cancer Nanotechnology Excellence Grant U54 CA119367
(Box 1). The pharmacokinetics and toxicity of the NIRF (to S.S.G.).
nanoprobes should be investigated in parallel with their
imaging potential. Food and Drug Administration ap- References
proved, biodegradable and biocompatible materials might 1 Massoud, T.F. and Gambhir, S.S. (2003) Molecular imaging in living
subjects: seeing fundamental biological processes in a new light. Genes
be good starting materials for constructing the NIRF Dev. 17, 545–580
nanoprobes. Although there are examples for successful 2 Weissleder, R. (2006) Molecular imaging in cancer. Science 312, 1168–
imaging of tumors in small animal models using NIRF 1171
nanoprobes, an unfavorable biodistribution such as highly 3 Quon, A. and Gambhir, S.S. (2005) FDG-PET and beyond: molecular
nonspecific accumulation in the RES still needs to be breast cancer imaging. J. Clin. Oncol. 23, 1664–1673
4 Weissleder, R. and Pittet, M.J. (2008) Imaging in the era of molecular
resolved. The major challenge is to develop the surface oncology. Nature 452, 580–589
5 Willmann, J.K. et al. (2008) Molecular imaging in drug development.
Nat. Rev. Drug Discov. 7, 591–607
Box 1. Aspects of nanoprobes that need to be addressed 6 Cai, W.B. et al. (2006) PET of vascular endothelial growth factor
before clinical translation will be possible receptor expression. J. Nucl. Med. 47, 2048–2056
7 Holman, B.L. and Devous, M.D. (1992) Functional brain SPECT – the
 Biodegradable and biocompatible materials should be considered emergence of a powerful clinical method. J. Nucl. Med. 33, 1888–1904
as starting materials for constructing the NIRF nanoprobes. 8 Harisinghani, M.G. et al. (2003) Noninvasive detection of clinically
 Synthesis of novel NIRF nanoprobes with high quantum yield and occult lymph-node metastases in prostate cancer. N. Engl. J. Med.
deep tissue penetration. 348, 2491–2499
 Sufficient photostability for applications in living subjects. 9 McCarthy, J.R. and Weissleder, R. (2008) Multifunctional magnetic
 Appropriate surface coating for decreasing nonspecific binding nanoparticles for targeted imaging and therapy. Adv. Drug Deliv. Rev.
while in circulation or during RES uptake. 60, 1241–1251
 A reasonable size of NIRF nanoprobes to allow extravasation and 10 Gao, J.H. et al. (2009) Multifunctional magnetic nanoparticles: design,
a half-life long enough for extravasation to occur. synthesis, and biomedical applications. Acc. Chem. Res. 42, 1097–1107
 Development of targeted NIRF nanoprobes with high specificity 11 Ballou, B. et al. (2005) Fluorescence imaging of tumors in vivo. Curr.
and selectivity. Med. Chem. 12, 795–805
 Development of multimodal and multifunctional nanoprobes for 12 Hilderbrand, S.A. and Weissleder, R. (2010) Near-infrared
several applications. fluorescence: application to in vivo molecular imaging. Curr. Opin.
Chem. Biol. 14, 71–79

581
Review Trends in Molecular Medicine Vol.16 No.12

13 Gao, J.H. et al. (2010) Near-infrared quantum dots as optical probes 42 Sun, Y.P. et al. (2006) Quantum-sized carbon dots for bright and
for tumor imaging. Curr. Top. Med. Chem. 10, 1147–1157 colorful photoluminescence. J. Am. Chem. Soc. 128, 7756–7757
14 Minchin, R.F. and Martin, D.J. (2010) Nanoparticles for molecular 43 Park, J.H. et al. (2009) Biodegradable luminescent porous silicon
imaging – an overview. Endocrinology 151, 474–481 nanoparticles for in vivo applications. Nat. Mater. 8, 331–336
15 Yang, X. et al. (2009) Nanoparticles for photoacoustic imaging. Wiley 44 Burns, A.A. et al. (2009) Fluorescent silica nanoparticles with efficient
Interdiscip. Rev. Nanomed. Nanobiotechnol. 1, 360–368 urinary excretion for nanomedicine. Nano Lett. 9, 442–448
16 De la Zerda, A. et al. (2008) Carbon nanotubes as photoacoustic 45 De, A. and Gambhir, S.S. (2005) Noninvasive imaging of protein–
molecular imaging agents in living mice. Nat. Nanotechnol. 3, 557–562 protein interactions from live cells and living subjects using
17 Lee, J.H. et al. (2007) Artificially engineered magnetic nanoparticles bioluminescence resonance energy transfer. FASEB J. 19, 2017
for ultra-sensitive molecular imaging. Nat. Med. 13, 95–99 46 De, A. et al. (2009) BRET3: a red-shifted bioluminescence resonance
18 He, X.X. et al. (2010) In vivo near-infrared fluorescence imaging of energy transfer (BRET)-based integrated platform for imaging
cancer with nanoparticles-based probes. Wiley Interdiscip. Rev. protein–protein interactions from single live cells and living
Nanomed. Nanobiotechnol. 2, 349–366 animals. FASEB J. 23, 2702–2709
19 Vetrone, F. and Capobianco, J.A. (2008) Lanthanide-doped fluoride 47 So, M.K. et al. (2006) Self-illuminating quantum dot conjugates for in
nanoparticles: luminescence, upconversion, and biological vivo imaging. Nat. Biotechnol. 24, 339–343
applications. Int. J. Nanotechnol. 5, 1306–1339 48 Zhang, Y. et al. (2006) HaloTag protein-mediated site-specific
20 Altinoglu, E.I. et al. (2008) Near-infrared emitting fluorophore-doped conjugation of bioluminescent proteins to quantum dots. Angew.
calcium phosphate nanoparticles for in vivo imaging of human breast Chem. Int. Ed. 45, 4936–4940
cancer. ACS Nano 2, 2075–2084 49 Xia, Z.Y. and Rao, J.H. (2009) Biosensing and imaging based on
21 Lee, C.H. et al. (2009) Near-infrared mesoporous silica nanoparticles bioluminescence resonance energy transfer. Curr. Opin. Biotechnol.
for optical imaging: characterization and in vivo biodistribution. Adv. 20, 37–44
Funct. Mater. 19, 215–222 50 Loening, A.M. et al. (2007) Red-shifted Renilla reniformis luciferase
22 Tanisaka, H. et al. (2008) Near-infrared fluorescent labeled variants for imaging in living subjects. Nat. Methods 4, 641–643
peptosome for application to cancer imaging. Bioconjug. Chem. 19, 51 Liu, H.G. et al. (2010) Radiation-luminescence-excited quantum dots
109–117 for in vivo multiplexed optical imaging. Small 6, 1087–1091
23 Cao, W.G. et al. (2009) Synthesis and evaluation of a stable 52 O’Connell, M.J. et al. (2002) Band gap fluorescence from individual
bacteriochlorophyll-analog and its incorporation into high-density single-walled carbon nanotubes. Science 297, 593–596
lipoprotein nanoparticles for tumor imaging. Bioconjug. Chem. 20, 53 Amiot, C.L. et al. (2008) Near-infrared fluorescent materials for
2023–2031 sensing of biological targets. Sensors 8, 3082–3105
24 Mulder, W.J.M. et al. (2009) Nanoparticulate assemblies of 54 Hong, H. et al. (2009) Molecular imaging with single-walled carbon
amphiphiles and diagnostically active materials for multimodality nanotubes. Nano Today 4, 252–261
imaging. Acc. Chem. Res. 42, 904–914 55 Liu, Z. et al. (2009) Carbon nanotubes in biology and medicine: in vitro
25 Yang, Z. et al. (2007) Long-circulating near-infrared fluorescence core- and in vivo detection, imaging and drug delivery. Nano Res. 2, 85–120
cross-linked polymeric micelles: synthesis, characterization, and dual 56 Huang, C.C. et al. (2008) Bioconjugated gold nanodots and
nuclear/optical imaging. Biomacromolecules 8, 3422–3428 nanoparticles for protein assays based on photoluminescence
26 Yang, Z. et al. (2009) Pharmacokinetics and biodistribution of near- quenching. Anal. Chem. 80, 1497–1504
infrared fluorescence polymeric nanoparticles. Nanotechnology 20, 57 Qian, X.M. et al. (2008) Surface-enhanced raman nanoparticle
165101 beacons based on bioconjugated gold nanocrystals and long range
27 Bruchez, M. et al. (1998) Semiconductor nanocrystals as fluorescent plasmonic coupling. J. Am. Chem. Soc. 130, 14934–14935
biological labels. Science 281, 2013–2016 58 Niidome, T. et al. (2008) In vivo monitoring of intravenously injected
28 Chan, W.C.W. and Nie, S.M. (1998) Quantum dot bioconjugates for gold nanorods using near-infrared light. Small 4, 1001–1007
ultrasensitive nonisotopic detection. Science 281, 2016–2018 59 Keren, S. et al. (2008) Noninvasive molecular imaging of small living
29 Michalet, X. et al. (2005) Quantum dots for live cells, in vivo imaging, subjects using Raman spectroscopy. Proc. Natl. Acad. Sci. U. S. A. 105,
and diagnostics. Science 307, 538–544 5844–5849
30 Medintz, I.L. et al. (2003) Self-assembled nanoscale biosensors based 60 Zheng, J. et al. (2007) Highly fluorescent noble-metal quantum dots.
on quantum dot FRET donors. Nat. Mater. 2, 630–638 Annu. Rev. Phys. Chem. 58, 409–431
31 Gill, R. et al. (2008) Semiconductor quantum dots for bioanalysis. 61 Zheng, J. et al. (2003) High quantum yield blue emission from water-
Angew. Chem. Int. Ed. 47, 7602–7625 soluble Au-8 nanodots. J. Am. Chem. Soc. 125, 7780–7781
32 Smith, A.M. et al. (2008) Bioconjugated quantum dots for in vivo 62 Bao, Y.P. et al. (2007) Nanoparticle-free synthesis of fluorescent gold
molecular and cellular imaging. Adv. Drug Deliv. Rev. 60, 1226–1240 nanoclusters at physiological temperature. J. Phys. Chem. C 111,
33 Frangioni, J.V. (2003) In vivo near-infrared fluorescence imaging. 12194–12198
Curr. Opin. Chem. Biol. 7, 626–634 63 Huang, C.C. et al. (2009) Synthesis of wavelength-tunable
34 Tsay, J.M. et al. (2004) Hybrid approach to the synthesis of highly luminescent gold and gold/silver nanodots. J. Mater. Chem. 19,
luminescent CdTe/ZnS and CdHgTe/ZnS nanocrystals. J. Am. Chem. 755–759
Soc. 126, 1926–1927 64 Xie, J.P. et al. (2009) Protein-directed synthesis of highly fluorescent
35 Kim, S. et al. (2004) Near-infrared fluorescent type II quantum dots gold nanoclusters. J. Am. Chem. Soc. 131, 888–889
for sentinel lymph node mapping. Nat. Biotechnol. 22, 93–97 65 Dass, C.R. (2004) Tumour angiogenesis, vascular biology and
36 Kim, S.W. et al. (2005) Engineering InAsxP1-x/InP/ZnSe III-V alloyed enhanced drug delivery. J. Drug Target. 12, 245–255
core/shell quantum dots for the near-infrared. J. Am. Chem. Soc. 127, 66 Davis, M.E. et al. (2008) Nanoparticle therapeutics: an emerging
10526–10532 treatment modality for cancer. Nat. Rev. Drug Discov. 7, 771–782
37 Allen, P.M. and Bawendi, M.G. (2008) Ternary I III VI quantum 67 Choi, H.S. et al. (2007) Renal clearance of quantum dots. Nat.
dots luminescent in the red to near-infrared. J. Am. Chem. Soc. 130, Biotechnol. 25, 1165–1170
9240–9241 68 Choi, H.S. et al. (2010) Design considerations for tumour-targeted
38 Xie, R.G. and Peng, X.G. (2009) Synthesis of Cu-doped InP nanoparticles. Nat. Nanotechnol. 5, 42–47
nanocrystals (d-dots) with ZnSe diffusion barrier as efficient and 69 Gao, J.H. et al. (2010) Ultrasmall near-infrared non-cadmium
color-tunable NIR emitters. J. Am. Chem. Soc. 131, 10645–10651 quantum dots for in vivo tumor imaging. Small 6, 256–261
39 Derfus, A.M. et al. (2004) Probing the cytotoxicity of semiconductor 70 Hama, Y. et al. (2007) Simultaneous two-color spectral fluorescence
quantum dots. Nano Lett. 4, 11–18 lymphangiography with near infrared quantum dots to map two
40 Kirchner, C. et al. (2005) Cytotoxicity of colloidal CdSe and CdSe/ZnS lymphatic flows from the breast and the upper extremity. Breast
nanoparticles. Nano Lett. 5, 331–338 Cancer Res. Treat. 103, 23–28
41 Xie, R.G. et al. (2009) Formation of high-quality I-III-VI 71 Welsher, K. et al. (2009) A route to brightly fluorescent carbon
semiconductor nanocrystals by tuning relative reactivity of cationic nanotubes for near-infrared imaging in mice. Nat. Nanotechnol. 4,
precursors. J. Am. Chem. Soc. 131, 5691–5697 773–780

582
Review Trends in Molecular Medicine Vol.16 No.12

72 Gao, X.H. et al. (2004) In vivo cancer targeting and imaging with 88 Park, J.H. et al. (2008) Micellar hybrid nanoparticles for simultaneous
semiconductor quantum dots. Nat. Biotechnol. 22, 969–976 magnetofluorescent imaging and drug delivery. Angew. Chem. Int.
73 Cai, W.B. et al. (2006) Peptide-labeled near-infrared quantum dots for Ed. 47, 7284–7288
imaging tumor vasculature in living subjects. Nano Lett. 6, 669–676 89 Cai, W.B. et al. (2007) Dual-function probe for PET and near-infrared
74 Cai, W.B. and Chen, X.Y. (2008) Preparation of peptide-conjugated fluorescence imaging of tumor vasculature. J. Nucl. Med. 48, 1862–1870
quantum dots for tumor vasculature-targeted imaging. Nat. Protoc. 3, 90 Chen, K. et al. (2008) Dual-modality optical and positron emission
89–96 tomography imaging of vascular endothelial growth factor receptor on
75 Gao, J.H. et al. (2010) In vivo tumor-targeted fluorescence imaging tumor vasculature using quantum dots. Eur. J. Nucl. Med. Mol.
using near-infrared non-cadmium quantum dots. Bioconjug. Chem. Imaging 35, 2235–2244
21, 604–609 91 Nahrendorf, M. et al. (2010) Hybrid PET-optical imaging using
76 Smith, B.R. et al. (2008) Real-time intravital imaging of RGD- targeted probes. Proc. Natl. Acad. Sci. U. S. A. 107, 7910–7915
quantum dot binding to luminal endothelium in mouse tumor 92 Schipper, M.L. et al. (2007) MicroPET-based biodistribution of
neovasculature. Nano Lett. 8, 2599–2606 quantum dots in living mice. J. Nucl. Med. 48, 1511–1518
77 Schellenberger, E. (2010) Bioresponsive nanosensors in medical 93 Schipper, M.L. et al. (2009) Particle size, surface coating, and
imaging. J. R. Soc. Interface 7, S83–91 PEGylation influence the biodistribution of quantum dots in living
78 Jiang, T. et al. (2004) Tumor imaging by means of proteolytic mice. Small 5, 126–134
activation of cell-penetrating peptides. Proc. Natl. Acad. Sci. U. S. A. 94 Storm, G. et al. (1995) Surface modification of nanoparticles to oppose
101, 17867–17872 uptake by the mononuclear phagocyte system. Adv. Drug Deliv. Rev.
79 Lee, S. et al. (2008) Activatable imaging probes with amplified 17, 31–48
fluorescent signals. Chem. Commun. 4250–4260 95 Aillon, K.L. et al. (2009) Effects of nanomaterial physicochemical
80 Lee, S. et al. (2008) A near-infrared-fluorescence-quenched gold- properties on in vivo toxicity. Adv. Drug Deliv. Rev. 61, 457–466
nanoparticle imaging probe for in vivo drug screening and 96 Lewinski, N. et al. (2008) Cytotoxicity of nanoparticles. Small 4, 26–49
protease activity determination. Angew. Chem. Int. Ed. 47, 2804–2807 97 Hauck, T.S. et al. (2010) In vivo quantum-dot toxicity assessment.
81 Lee, S. et al. (2010) Activatable molecular probes for cancer imaging. Small 6, 138–144
Curr. Top. Med. Chem. 10, 1135–1144 98 Schipper, M.L. et al. (2008) A pilot toxicology study of single-walled
82 Chang, E. et al. (2005) Protease-activated quantum dot probes. carbon nanotubes in a small sample of mice. Nat. Nanotechnol. 3, 216–
Biochem. Biophys. Res. Commun. 334, 1317–1321 221
83 Cai, W.B. and Chen, X.Y. (2007) Nanoplatforms for targeted 99 Hardman, R. (2006) A toxicologic review of quantum dots: toxicity
molecular imaging in living subjects. Small 3, 1840–1854 depends on physicochemical and environmental factors. Environ.
84 Mulder, W.J.M. et al. (2007) Magnetic and fluorescent nanoparticles Health Perspect. 114, 165–172
for multimodality imaging. Nanomedicine 2, 307–324 100 Nel, A. et al. (2006) Toxic potential of materials at the nanolevel.
85 Cai, W.B. and Chen, X.Y. (2008) Multimodality molecular imaging of Science 311, 622–627
tumor angiogenesis. J. Nucl. Med. 49, 113–128S 101 Nguyen, Q.T. et al. (2010) Surgery with molecular fluorescence
86 Josephson, L. et al. (2002) Near-infrared fluorescent nanoparticles imaging using activatable cell-penetrating peptides decreases
as combined MR/optical imaging probes. Bioconjug. Chem. 13, 554– residual cancer and improves survival. Proc. Natl. Acad. Sci. U. S. A.
560 107, 4317–4322
87 Ostendorp, M. et al. (2008) Quantitative molecular magnetic 102 Olson, E.S. et al. (2010) Activatable cell penetrating peptides linked to
resonance imaging of tumor angiogenesis using cNGR-labeled nanoparticles as dual probes for in vivo fluorescence and MR imaging
paramagnetic quantum dots. Cancer Res. 68, 7676–7683 of proteases. Proc. Natl. Acad. Sci. U. S. A. 107, 4311–4316

583

Вам также может понравиться