You are on page 1of 13

Advanced Drug Delivery Reviews 65 (2013) 1951–1963

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


journal homepage: www.elsevier.com/locate/addr

Carbon nanotubes for biomedical imaging: The recent advances☆


Hua Gong, Rui Peng ⁎, Zhuang Liu ⁎
Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China

a r t i c l e i n f o a b s t r a c t

Article history: This article reviews the latest progresses regarding the applications of carbon nanotubes (CNTs), including single-
Accepted 16 October 2013 walled carbon nanotubes (SWNTs) and multi-walled carbon nanotubes (MWNTs), as multifunctional nano-
Available online 30 October 2013 probes for biomedical imaging. Utilizing the intrinsic band-gap fluorescence of semi-conducting single-walled
carbon nanotubes (SWNTs), fluorescence imaging in the near infrared II (NIR-II) region with enhanced tissue
Keywords:
penetration and spatial resolution has shown great promise in recent years. Raman imaging based on the reso-
Carbon nanotubes
Biomedical imaging
nance Raman scattering of SWNTs has also been explored by a number of groups for in vitro and in vivo imaging
NIR-II fluorescence of biological samples. The strong absorbance of CNTs in the NIR region can be used for photoacoustic imaging, and
Raman scattering their photoacoustic signals can be dramatically enhanced by adding organic dyes, or coating with gold shells. Tak-
Photoacoustic ing advantages of metal nanoparticle impurities attached to nanotubes, CNTs can also serve as a T2-contrast
Magnetic resonance agent in magnetic resonance (MR) imaging. In addition, when labeled with radioactive isotopes, many groups
Nuclear imaging have developed nuclear imaging with functionalized CNTs. Therefore CNTs are unique imaging probes with
great potential in biomedical multimodal imaging.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1951
2. Fluorescence imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1952
3. Raman imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1953
4. Photoacoustic imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1957
5. Magnetic resonance imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1958
6. Nuclear imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1959
7. Prospects and challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1960
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1960
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1960

1. Introduction

Carbon nanotubes (CNTs), including single-walled carbon nano-


tubes (SWNTs) and multi-walled carbon nanotubes (MWNTs), have be-
Abbreviations: CNTs, carbon nanotubes; SWNTs, single-walled carbon nanotubes; NIR,
near infrared; MR, magnetic resonance; 1D, one dimensional; PA, photoacoustic; PET, posi-
come star materials since its first discovery in 1990 [1–3]. Various
tron emission tomography; SPECT, single photon emission computed tomography; SERS, sur- applications related to CNTs, such as composite materials [4], nano-
face enhanced Raman scattering; QDs, quantum dots; DOS, density of states; VHSs, van Hoff electronics [5], field effect emitters [6], as well as in the area of energy
singularities; PEG, polyethylene glycol; QY, quantum yield; PL–PEG, phospholipid–PEG; PCA, research [7], have been extensively explored due to the unique physical
principle component analysis; C18PMH-PEG, poly(maleic anhydride-alt-1-octadecene)-
and chemical properties of this type of one-dimensional nanomaterials.
polyethylene glycol; EPR, enhanced permeability and retention; RBM, radical breathing
mode; PAH, poly(allylamine hydrochloride); GNTs, golden nanotubes; CTCs, circulating In the area of biomedicine, CNTs have also received tremendous atten-
tumor cells; ICG, indocyanine green; US-tubes, ultra-short SWNTs; MSC, mesenchymal tions. A large variety of CNT-based bio-sensors with different sensing
stem cells; hMSCs, human MSCs; DOTA, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic mechanisms have been reported by numerous groups for detections
acid. of different biological molecules [8,9]. Drug delivery and cancer therapy
☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Carbon
nanotubes in medicine and biology — Therapy and diagnostics”.
with CNTs are another intensively explored field attracting great inter-
⁎ Corresponding authors. ests [10–18]. In the direction of bio-imaging, CNT-based imaging probes
E-mail addresses: rpeng@suda.edu.cn (R. Peng), zliu@suda.edu.cn (Z. Liu). have also been widely investigated [19–24].

0169-409X/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.addr.2013.10.002
1952 H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963

The unique physical properties of CNTs, particularly SWNTs, make fluorescence imaging with improved tissue penetration and much
them extremely attractive in the area of biomedical imaging. The better spatial resolutions [26,43,57–59].
quasi one-dimensional (1D) semi-conducting SWNTs exhibit a narrow SWNTs are quasi one dimensional quantum wires with energy levels
band gap of about 1 eV [25], which allows fluorescence emission in become split as a result of quantum confinement effect. The density of
the near infrared (NIR) regions, including the classical NIR-I region states (DOS) are characterized by the so called van Hoff singularities
(700–900 nm) and the newly defined NIR-II region (1100–1400 nm) (VHSs), which define narrow energy ranges where the DOS intensity
[26–28]. SWNTs possess strong resonance Raman scattering with ex- is very high [60,61]. The band-gap between each semiconducting
tremely large scattering cross-section, and are thus great Raman probes SWNTs is in the order of 1 eV, which allows for the fluorescence in the
useful in biological sensing and imaging [22,29]. CNTs as one of the NIR-II region under the excitation in the NIR-I region [61]. (Fig. 1a&c)
darkest materials exhibit strong absorbance in the NIR region, and Furthermore, the large Stoke-shift between the excitation at 550–
therefore can be used as photoacoustic (PA) imaging contrast agents 850 nm and emission at 900–1600 nm (Fig. 1c) would dramatically
[30,31]. The impurities of metal nanoparticles contained in the CNT lower the autofluorescence of biological tissues during imaging, offering
samples can be utilized in magnetic resonance (MR) imaging, offering enhanced in vivo imaging sensitivity. (Fig. 1d–g) [28].
strong T2-weighted imaging contrast [32]. Besides adopting the intrin- Despite the encouraging results for the use of SWNTs in NIR fluores-
sic characteristics of CNTs, radionuclides, can also be conjugated or cence imaging, the low quantum yield (QY) of SWNTs at that time was
even inserted to CNTs to render more modalities of imaging, including the major limitation for the further applications of SWNTs for in vivo
positron emission tomography (PET) [33] and single photon emission imaging. Covalent functionalization would disrupt the structure of
computed tomography (SPECT) [34]. SWNTs and lead to complete elimination of their NIR fluorescence.
In the past several years, several review papers have summarized SWNTs suspended by small surfactant molecules such as sodium cho-
the progresses of using SWNTs in the area of biomedicine. Wu et al. late showed relatively high QY, however appeared to be toxic in biolog-
summarized different surface modification strategies to obtain func- ical systems. Non-covalent PEGylation of SWNTs, although can offer
tionalized SWNTs for biomedical use [35]. Kostarelos et al. discussed nanotubes great water-solubility and enhanced biocompatibility, usual-
the advantages and challenges of using CNTs in biomedicine in their ly would also drastically decrease of QY of SWNTs. In 2009, an interest-
progress article in 2009 [36]. In the same year, Liu et al. published a com- ing coating exchange method was developed by Dai and co-workers to
prehensive review which covers the surface modification, potential obtain biocompatible SWNTs with high QY. In their method, SWNTs
toxicity, as well as both in vitro and in vivo applications of CNTs [31]. were debundled and solubilized in a solution of sodium cholate, which
In 2010, we wrote another specific review paper about the applications was then displaced by phospholipid–PEG (PL–PEG). Compared with
of SWNTs in biomedical imaging [37]. However, considering tremen- the traditional modification strategy in which SWNTs were directly son-
dous progresses made in the very recent few years to explore CNT- icated in PL–PEG solutions over a long period (N 15 min), this relative
based biomedical imaging, an up-to-date review article is therefore gentle coating exchange method prevents the loss of QY for SWNTs.
needed to summarize the latest exciting results in this area. Using PEGylated SWNTs with high QY prepared by this method,
In this current review article, we will summarize the very recent in vivo whole body NIR-II fluorescence imaging of mice with intrave-
progresses on the applications of CNTs as multimodal contrast agents nously (i.v.) injected SWNTs was realized for the first time. Moreover,
for biomedical imaging. For example, in the past several years, the high-resolution in vivo intravital microscopy imaging was also realized
in vivo fluorescence imaging of SWNTs in the NIR-II region has made re- upon injection of SWNTs, visualizing small tumor vessels beneath the
markable progresses, making it an encouraging imaging tool with great thick skin [20].
potential in biomedical research [26,38–47]. By growing noble metal In a later work by the same group, Welsher et al. used SWNTs as NIR-
nanoparticles on SWNTs, their Raman scattering signals can be dramat- II contrast agent to perform the high frame rate fluorescent video imag-
ically enhanced by surface enhanced Raman scattering (SERS), allowing ing of mice i.v. injected with SWNTs, and investigated the path of
Raman imaging of biological samples with much faster speeds [48]. By SWNTs through mouse anatomy [26]. As shown in Fig. 2a, they observed
loading different dyes with NIR absorbance at varied wavelengths, in real time that SWNTs first reached the lungs several seconds after in-
multiplexed photoacoustic imaging in vivo has been demonstrated jection, and then the spleen and the liver at later time points. By means
[49]. Moreover, SWNTs with appropriate surface functionalization of principle component analysis (PCA), the anatomic resolution of
have also been used for stem cell labeling and in vivo multimodal organs was dramatically enhanced. Even the pancreas, which could
tracking based on Raman imaging, magnetic resonance imaging, not be resolved from real time raw images, could be discriminated
and photoacoustic imaging [32]. It is hoped that this review article after PCA analysis of NIR-II fluorescence images of mice with SWNT-
could offer a timely update regarding the use of CNTs in biomedical injection. (Fig. 2b) Therefore, this work demonstrated that the NIR-II
imaging. fluorescence imaging, together with PCA, could provide powerful tools
for a wide range of potential applications from biomedical research to
2. Fluorescence imaging disease diagnosis.
Subsequently, by making use of a new type of PEGylated amphiphilic
Fluorescence imaging plays a pivotal role in the scientific research polymer, poly(maleic anhydride-alt-1-octadecene)-polyethylene glycol
and medical diagnosis. However, the limited penetration depth of (C18PMH-PEG), to solubilize SWNTs, Robinson et al. prepared a well
light becomes the major hurdle for the further application of fluores- functionalized SWNTs formulation with long blood circulation (half-life
cence imaging [50]. To overcome this problem, researchers have been of 30 h) in vivo to achieve ultrahigh accumulation of ~30% inject dose
focusing on developing and implementing fluorescent probes with exci- (%ID/g) in 4T1 murine breast cancer bearing Balb/c mice [43]. For the
tation and emission wavelengths falling into the biological transparent first time, high fluorescent video rate imaging and PCA were adopted to
NIR window [51]. The classical NIR window, or NIR I window, is defined monitor the fluorescent signals in tumors and other organs. They found
to be from ~700 nm to ~900 nm, within which neither hemoglobin nor obvious fluorescent signals in the tumor only 20 s after injection, and
water exhibits significant light absorbance. Currently, various probes, the fluorescent signal remained there for up to 72 h. Furthermore, the
including organic dyes [52–54], as well as semiconducting quantum 3D reconstruction of NIR-II fluorescence signals inside tumors revealed
dots (QDs) [55] lie within this region. Recently, it has been uncovered the co-localization of tumor vasculatures and SWNTs, indicating that
that light with even longer wavelength in the range of 1100– the enhanced permeability and retention (EPR) effect may serve as
1400 nm, although would be slightly absorbed by water, shows further important role in mediating the tumor accumulation of nanotubes.
reduced tissue absorbance, and more importantly, remarkably reduced Besides tumor imaging, high fluorescent video rate imaging and PCA
scattering by biological tissues, allowing in vitro assay [56] and in vivo analysis were also adopted for vessel imaging in vivo. In a recent work
H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963 1953

Fig. 1. Comparison of NIR-I and NIR-II fluorescence imaging. IRDye800 conjugated SWNTs emit with both NIR-I (from IRDye 800) and NIR-II (from SWNTs) fluorescence were used in this
experiment. (a) A schematic showing that upon excitation by a 785 nm laser, the SWNTs-IRDye-800 conjugate emits at the 800 nm NIR-I region from IRDye-800 dye and the 1100–
1400 nm NIR-II region from SWNTs. (b) A schematic of the imaging setup for simultaneous detection of both NIR-I and NIR-II photons using silicon and InGaAs cameras.(c) Absorption
spectrum of the SWNTs–IRDye-800 conjugate (black dashed line) together with emission spectra of IRDye-800 (green line) and SWNTs (red line).(d–g) NIR-II (d) and NIR-I (f) fluorescent
images and the corresponding cross-section fluorescent intensity profiles (e & g) along red-dashed bars of a mouse injected with SWNTs–IRDye-800. Copyright 2012 Nature Publishing
Group [28].

by Hong et al., arterial and venous vessels were unambiguously differ- In a short summary, NIR-II fluorescent imaging with SWNTs has
entiated using a dynamic contrast-enhanced NIR-II imaging technique shown great potential in biomedical imaging, demonstrating several
on basis of their distinct hemodynamics. In Fig. 3a–d, the NIR-II image unique advantages over other existing imaging techniques. Further de-
showed greater numbers of small vessels in the distal hind limp of velopment in this direction requires better SWNT samples with high QY
mouse compared to micro-CT at the same location. The smallest vessel and purified chiralities to provide even ‘brighter’ fluorescence. More-
by NIR-II had a Gaussian-fit diameter of only 35.4 μm (Fig. 3g), where over, different SWNTs with purified single chiralities would show sepa-
micro-CT could not discern any vessels smaller than 100 μm (Fig. 3h). rated excitation and emission wavelengths, potentially useful for multi-
Furthermore, the blood velocity inside the vessels could also be mea- color NIR-II fluorescence imaging, which remains to be demonstrated in
sured in both ischemic and normal limbs, even at very low blood veloc- future studies. Other fluorescence enhancement techniques, such as
ities, which are beyond the capabilities of ultrasonography. Therefore, gold substrate based surface resonance enhancement of SWNT fluores-
their results suggested that the NIR-II fluorescence imaging of vascula- cence [47], may also be integrated for further improved imaging and
tures using SWNTs as nano-probes showed several advantages when detection sensitivity.
compared with Micro-CT and ultrasound imaging techniques [28].
As-synthesized SWNTs currently in use all contain various chiral- 3. Raman imaging
ities, and each chirality corresponds to a different set of excitation and
emission wavelengths. Therefore, in previous NIR-II fluorescence imag- Unlike fluorescence, Raman scattering also involves emission of pho-
ing experiments, only a small portion of SWNTs are excited by the laser tons with shifted wavelengths under light excitation, is a photon scat-
to give fluorescence, while the major portion is not in the resonance tering process rather than photoluminescence. The inherent Raman
with the excitation laser wavelength would be ‘dark’ during imaging. scattering signals of molecules without involving enhancement mecha-
Therefore, if chirality-purified SWNTs are used for NIR-II fluorescence nism such as SERS are usually rather weak. However, the scattering ef-
imaging, the amount of nanotubes injected into animals would be re- ficiency gets larger when the laser energy matches the energy needed
markably reduced. Various strategies have been developed in order to for the electron transition from the valence to the conduction bands,
obtain SWNTs with pure chiralities, including dielectrophoresis, density and this enhancement of Raman signals is also named resonance
gradient centrifugation [62], DNA wrapping chromatography [63], and Raman scattering [67]. Typically, SWNTs have multiple Raman peaks,
gel filtration [64–66]. In a recent work, Diao et al. developed a simple including the radical breathing mode (RBM, 100 cm−1–300 cm−1)
gel separation method to enrich semiconducting (12,1) and (11,3) and tangential G band (~1580 cm−1), which correspond to the vibra-
SWNTs with identical resonance absorption at 808 nm and emission tion of carbon atoms in the radical direction and in the tangential direc-
at 1200 nm. The chirality sorted SWNTs exhibited 5 folds higher tion, respectively. The resonance Raman scattering for SWNTs is
fluorescent intensity under the resonant excitation at 808 nm than determined by the DOS available for the optical transition (e.g. E11 and
unsorted SWNTs on a per mass basis, dramatically lowering the in- E22 transition), which is heavily determined by their diameters and
jection dosage of SWNTs (~ 6 folds) needed for in vivo imaging [42]. chirality indices [68]. When the Raman spectrum of SWNTs is taken,
1954 H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963

Fig. 2. Video-rate NIR-II imaging of SWNTs in a live mouse. (a–h) Frames from video imaging of a mouse injected with PEGylated SWNTs taken at different time points post injection. (i).
Dynamic contrast-enhanced imaging of the mouse injected with SWNTs through PCA. PCA images taken over the first 130 s following injection were performed by taking every 150 evenly
spaced frames out of the 2000-frame dataset. Major features observed belong to the lungs, liver, kidney, spleen and even the pancreas. Copyright 2011 National Academy of Sciences [26].

those SWNTs with Eii in resonance would offer strong resonance Raman to recognize the corresponding cell lines. By means of spectrum
scattering useful for biological sensing and imaging. unmixing, multiplexed Raman imaging was realized simultaneously. A
The intrinsic Raman properties of SWNTs were first adopted by Heller later work by the same group went a further step to realize the five-
et al. for live cell imaging in 2005 [27]. In their work, SWNTs wrapped by color Raman imaging of cells by conjugating SWNTs with different
DNA oligonucleotide were used as markers for live cell tracking. Without targeting molecules, for both multiplexed in vitro cell imaging and
adding any extra fluorescent labels, the RBM peak of SWNTs was record- ex vivo tissue slice staining [78].
ed inside live 3T3 cells under the excitation of a 785-nm laser. Compared Although SWNTs are likely to exhibit the strongest inherent Raman
with organic dyes and quantum dots which could be gradually quenched signals among all single molecules, for biological samples labeled with
or photo-bleached over time, SWNTs showed rather robust Raman SWNTs, the acquisition time to get a appropriate Raman image is still
scattering signals without any significant quenching or photobleaching, long (hours for a image with 100×100 pixels). Since the acquisition
allowing them to be used for long-term tracking in biological samples time in Raman imaging is highly determined by the Raman signals of
over months [69]. Taking advantages of the inherent Raman G band of Raman tags, the best way to shorten the imaging time is to enhance
carbon nanotubes, Lamprecht et al. were able to map and track the intra- the Raman signals of those tags. Surface enhanced Raman scattering
cellular distribution of carbon nanotubes after targeted delivery to carci- (SERS) may serve as possible way to enhance Raman signals of
noma cells [70]. In another recent study, Kang et al. used high-speed SWNTs by coating noble metal nanoparticles on the nanotube surface
confocal Raman imaging to study the cellular uptake of SWNTs. In their [48,79–81]. Although the on-substrate deposition of noble metal nano-
work, movies of two cell-intrinsic and nine nanotube-derived Raman particles on SWNTs to achieve the SERS effect has been widely reported
signals in RAW 264.7 macrophage were taken to resolve SWNTs with by many different groups [81–83], and there has been different methods
different indices and aggregation states, as well as their positions inside to attach gold nanoparticles to covalently functionalized SWNTs
the cells. This work highlighted the advantages of Raman spectroscopy which usually showed rather low Raman signals due to the damage of
for molecular imaging of live cells [71]. In vivo Raman imaging of nanotube structure during functionalization (e.g. by oxidization)
tumors on mice has also been demonstrated by using targeting ligand- [84,85], the development of noble metal coated pristine SWNTs with
conjugated SWNTs [72,73]. noncovalent functionalization as a SERS nano-probe for Raman imaging
Raman scattering usually exhibits rather sharp peaks with varied lo- of biological samples was not reported until a recent work by our group
cations for different molecules, and thus is ideal for multiplexed sensing [48]. In this article, single strand DNA was used to functionalize pristine
and imaging [74–76]. The frequency of carbon–carbon bond vibration in SWNTs with retained strong Raman scattering. After coating with posi-
the G band Raman peak of SWNTs is determined by the carbon atom tively charged poly(allylamine hydrochloride) (PAH), those nanotubes
mass. By changing the carbon isotope from 12C to 13C, the G band were then attached with negatively charged gold nano-seeds, which in-
peaks of SWNTs could be altered accordingly [77]. The first multiplexed duced the growth of gold or silver nano-shells on the surface of SWNTs.
Raman imaging of live cells was identified by Liu et al. [22]. In this paper, (Fig. 4a–d) The noble metal coated SWNTs gained excellent SERS effects,
three types SWNTs with varied 13C doping ratio and thus different with the maximal enhancement factor of over 20 (Fig. 4e–g) in the
Raman G-band peaks were conjugated with different targeting ligands solution phase. Owing to the markedly enhanced Raman scattering,
H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963 1955

Fig. 3. The comparison of SWNT-based NIR-II fluorescence imaging and micro-CT imaging. (a) A NIR-II SWNT fluorescence image of a mouse thigh. (b) A micro-CT image showing the same
area of the thigh as that in (a). (c) A cross-sectional fluorescence intensity profile measured along the green dashed line in (a) with its two peaks fitted to Gaussian functions. (d) A cross-
sectional intensity profile measured along the green dashed line in (b) with its two peaks fitted to Gaussian functions. (e) A NIR-II image at the level of the gastrocnemius. (f) A micro-CT
image showing the same area of the limb as in e. (g) A cross-sectional fluorescence intensity profile measured along the green dashed line in e with its peak fitted to a Gaussian function. (h)
A cross-sectional intensity profile measured along the green dashed line in (f) with its peak fitted to a Gaussian function. All scale bars indicate 2 mm. Copyright 2012 Nature Publishing
Group [28].

gold-coated SWNTs were then conjugated with folic acid for specific cell SWNTs as a novel Raman tag exhibit several advantages over
labeling and Raman imaging, whose imaging time was shortened by other organic Raman dyes. Firstly, the Raman peaks of SWNTs are
one order of magnitude to enable fast mapping of biological samples simple, narrow, and intense, with a full width at half-maximum
(Fig. 4h). less than 2 nm, which is very easy to be distinguished from the
1956 H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963

Fig. 4. Noble metal enhanced SERS of SWNTs. (a) Schematic illustration of the synthetic procedure of SWNT–Au (or SWNT–Ag) nanocomposite. (b) A SEM image of SWNT–Au–PEG nano-
composite. (c,d) TEM images of SWNT–Au–PEG nanocomposite. (e) A photo of SWNT–Au–PEG solutions prepared by adding different concentrations of growth solution. (f) Raman spectra
of SWNT–Au–PEG with different concentrations of growth solutions added during sample preparation. The spectra were taken under 785 nm laser excitation. (g) Enhanced factors of
SWNT–Au–PEG as a function of added gold growth solution concentrations under the 633-nm and 785-nm excitations. (h) High-resolution Raman image of a SWNT–Au–PEG–FA labeled
KB cell. The inset was a bright field image of this cell. Copyright 2012 American Chemical Society [48].

autofluorescent background. Secondly, the Raman signals of SWNTs multiple ‘colors’, allowing multiplexed Raman imaging. Lastly, the
are rather robust, without quenching or bleaching, enabling long- resonance Raman signals of SWNTs can be combined with the SERS
term tracking and imaging. Thirdly, the Raman shifts can be modu- technique, which further increases the detection sensitivity and re-
lated by changing the isotope composition within SWNTs to achieve duces the imaging time.
H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963 1957

4. Photoacoustic imaging In order to further enhance the sensitivity of photoacoustic signal of


SWNTs, a gold layer or some organic molecules with NIR absorbance
Photoacoustic (PA) imaging is a recently developed imaging modal- were coupled with SWNTs to increase their absorbance in the NIR re-
ity, which has been widely used in the biological field [86–88]. The prin- gion. Zharov and coworkers developed ‘golden nanotubes’ (GNTs) by
ciple of PA imaging is as follows: the laser pulses can be absorbed by coating CNTs with gold to enhance the intrinsic PA signals of SWNTs
light-absorbing molecules, either endogenous molecules or contrast [21]. In their work, the thin layer of gold encapsulated on the surface
agents, in the biological sample to generate heat, inducing transient of SWNTs enhanced their optical density at the NIR region, and then
thermoelastic expansion and thus leading to wideband ultrasonic emis- the GNTs were conjugated with antibodies to specifically recognize
sion, which can be detected by an ultrasound microphone to construct the endothelium of murine lymphatic vessels (Fig. 5a). The obtained
2D or 3D images. Compared with traditional optical imaging, PA imag- GNTs offered enhanced NIR PA imaging contrast (~102 fold) for
ing detects sound instead of light, and thus shows a number of obvious targeting lymphatic vessels in mice using extremely low laser fluency
advantages such as greatly improved tissue penetration and enhanced level at a few mJ/cm−2. Furthermore, antibody conjugated GNTs were
spatial resolutions, by avoiding absorbance and scattering of the emis- used to map the lymphatic endothelia receptor. As can be seen from
sion light in fluorescence imaging, respectively. Various nanomaterials Fig. 5, both the photoacoustic signal and photothermal signal in the an-
which have strong absorbance in the NIR region are useful contrast tibody conjugated group (Fig. 5c&g) exceeded that of endogenous back-
agents in PA imaging [89–91]. ground and were preferentially located in the wall of lymphatic vessels,
Both MWNTs and SWNTs have been adopted as a photothermal while for the GNTs without conjugation with antibodies (Fig. 5d,e,h&i),
agent due to its strong NIR absorbance [92–94]. On the other hand, its only random signals were seen and no signals could be found in the
strong NIR absorbance also makes nanotubes ideal contrast agents for lymphatic wall of vessels. A later work by the same group further ex-
photoacoustic imaging. For cell imaging, Avti et al. [95]. adopted photo- tended the application of GNTs to detect circulating tumor cells (CTCs)
acoustic microscopy to detect, map and quantify the trace amount under PA imaging. Taking advantage of the strong PA signals of GNTs,
(nanograms to micrograms) of SWNTs in a variety of histological tissue folate conjugated GNTs were used as the PA contrast agent to image
specimens. The results showed that the noise-equivalent detection sen- CTCs in vivo after being captured by an external magnetic field [96].
sitivity was as low as about 7 picogram, which allowed further applica- For the dye enhanced PA imaging with SWNTs, Gambhir et al. [49]
tion in tissue analysis. For in vivo PA imaging, Gambhir et al. for the first loaded indocyanine green (ICG) molecules on PEGylated SWNTs
time adopted RGD conjugated SWNTs as PA contrast agent. In their through pi–pi stacking, which increased the optical density of the
study, strong PA signals could be observed from the tumor in SWNT– nano-probe by ~20 fold at 780 nm. Compared to the sensitivity of PA
RGD injected group, while only weak signals were observed in the imaging obtained by plain SWNTs (~50 nM), SWNT–ICG exhibited
plain SWNTs injected group [19]. sub-nanomolar detection sensitivity in PA imaging. Based on the same

Fig. 5. In vivo molecular targeting of murine lymphatics with GNTs guided by an integrated photoacoustic (PA)/photothermal (PT) technique. (a) Schematics of GNTs synthesis and its
delivery to the target. (b–i) PT (b–e) and PA (f–i) two-dimensional lymphatic mapping in selected mesenteric areas taken before GNTs administration (b,f), at 60 min after administration
of antibody — GNTs (c,g), and at 15 min (d,h) and 60 min (e,i) after administration of GNTs alone. Dashed white lines in (b–i) indicated the lymphatic wall and valve. Copyright 2009
Nature Publishing Group [21].
1958 H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963

Fig. 6. Multiplexed photoacoustic imaging. (a) Optical absorbance spectra of SWNT–QSY (red), and SWNT–ICG (green). The QSY and ICG dye-enhanced SWNTs showed 17- and 20-times
higher optical absorption than plain SWNTs at their peak absorption wavelengths, 710 and 780 nm, respectively.(b) Optical absorbance spectra of plain SWNTs (black), SWNT-melanin
(purple), SWNT-Cy5.5 (brown), and SWNT-MB (blue). (c) PA imaging of mice subcutaneously injected with SWNT–QSY or SNWT–ICG at concentrations between 0.5 and 122 nM. PA im-
ages were acquired at λ = 710 nm (red) and 780 (green) excitation, for SWNT–QSY and SWNT–ICG, respectively. The skin and inclusion region were visualized in the ultrasound images
(black/white). (d) The in vivo PA signals from SWNT–ICG and SWNT–QSY at different SWNT concentrations. The error bars represented standard error (n = 3 mice). Linear regression
(R2 = 0.97 for both particles) of the two PA signal curves estimated that a concentration of 450 pM for SWNT–QSY or 170 pM for SWNT–ICG produced the equivalent background signal
of tissues. Copyright 2012 American Chemical Society [97].

concept, they further developed multiplexed PA imaging probes by For T2 weighted MR imaging, Strano and coworkers first reported
loading five types of NIR dyes with shifted absorbance peaks on that SWNTs with iron oxide nanoparticles attached at the end of nano-
PEGylated SWNTs (Fig. 6a&b) [97] In particular, SWNTs loaded with tubes could be used as T2 contrast agents for MR imaging, without the
QSY21 (SWNT–QSY) or indocyanine green (SWNT–ICG) exhibited need of additional labeling [110]. Wu et al. reported MWNTs/cobalt
strong and separated absorbance spectra, and could be used for multi- ferrite synthesized by a solvothermal method. The obtained nanocom-
color PA imaging in vivo with great sensitivity (Fig. 6c&d). posite exhibited a significant negative contrast enhancement in T2-
Therefore, CNTs with strong NIR absorbance are promising contrast weighted MR imaging of cancer cells [114].
agents in photoacoustic imaging. In addition to the use of their intrinsic Metal catalysts (e.g. Fe, Co) are commonly used during the synthesis
optical absorbance, CNTs could also serve as a versatile nano-platform of CNTs. Taking advantages of the impurities of iron contained in
by coupling them with other light-absorbing nano-structures or mole- SWNTs, Faraj et al. studied the biodistribution of SWNTs by measuring
cules for enhanced or multiplexed photoacoustic imaging. Although MR signals in vivo [109]. Even after purification to remove the majority
most of currently reported CNT-based photoacoustic imaging probes of catalyst metal nanoparticles in the nanotube sample, the purified
are based on SWNTs, MWNTs should also be useful for this type of SWNTs with trace amount of iron content have been found to be still
imaging modality. effective to offer strong T2-contrast in MR imaging [115].
Stem cells have shown great potential in regenerative medicine and
5. Magnetic resonance imaging attracted tremendous interests in recent years [116,117]. Sensitive and
reliable methods for stem cell labeling and in vivo tracking are thus
Magnetic resonance (MR) imaging is one of the most powerful and urgently needed. Carbon nanotubes have also been used for stem cell
noninvasive modalities of imaging [98]. Annually, about 60 million of labeling and tracking by several groups. Vittorrio et al. reported that
MR imaging cases are conducted in the clinic worldwide, and 30% of MWNTs labeled mesenchymal stem cells (MSC) could be successfully
them use contrast agents [99]. Generally, the contrast agents are consti- tracked after being administrated into animals under T2-weighted MR
tuted of T1-shortening agents (containing Gd3+, Mn2+, etc.) and T2- imaging [118]. Our group recently used SWNTs to label human MSCs
shortening agents (e.g. iron oxide nanoparticle). Considerable amount (hMSCs) for in vivo tracking based on triple modal imaging [32]. PEG
of research has been carried out to explore the potential application of functionalized SWNTs were conjugated with protamine, which remark-
CNTs for MR imaging [32,100–113]. ably enhanced the cellular uptake of SWNTs for highly efficient stem cell
For T1 weighted MR imaging, Sitharaman et al. first reported the labeling (Fig. 7a,c&d). SWNT-labeled hMSCs showed unaffected differ-
nanoscale loading and confinement of aquated Gd3+-ion clusters entiation and proliferation abilities compared to un-labeled cells. The
within ultra-short SWNTs (US-tubes), obtaining Gd3+@US-tube with strong inherent resonance Raman scattering of SWNTs was utilized for
strong T1-contrast useful in MR imaging [107]. Richard et al. reported in vitro and in vivo Raman imaging of SWNT-labeled hMSCs, enabling
a noncovalent strategy to conjugate MWNTs with an amphiphilic gado- ultrasensitive in vivo detection of as few as 500 stem cells administrated
linium (III) chelate (GdL), making them useful to produce T1-contrast in into a mouse. On the other hand, the metallic catalyst nanoparticles at-
MR imaging [108]. tached on nanotubes (Fig. 7b) can be utilized as the T2-contrast agent in
H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963 1959

Fig. 7. SWNTs used for stem cell tracking. (a) A schematic drawing of PEGylated SWNTs with protamine conjugation (SWNT–PEG–PRO). (b) A TEM images of SWNTs. (c,d) Raman images
of hMSCs incubated with SWNT–PEG (c) and SWNT–PEG–PRO(d). (e) In vivo T2-weighted MR image of a mouse injected with SWNT-labeled hMSCs. Arrows pointed to the sites where
unlabeled (left) and SWNT-labeled (right) hMSCs were injected. (f & g) In vivo Raman images of unlabeled (f) and SWNT-labeled (g) hMSCs. (h & i) In vivo PA images of unlabeled (h) and
SWNT-labeled (i) hMSCs. The circles highlighted the locations were hMSCs were injected. PA signals in the rectangle highlighted in h were from a blood vessel crossing this area. Copyright
2012 Wiley [32].

magnetic resonance (MR) imaging of SWNT-labeled hMSCs (Fig. 7e). 6. Nuclear imaging
Moreover, in vivo photoacoustic imaging of hMSCs in mice is also dem-
onstrated (Fig. 7h&i). This work reveals that SWNTs with appropriate Besides adopting the intrinsic characteristics of SWNTs, external la-
surface functionalization could serve as multifunctional nanoprobes bels such as radio-isotopes can also be introduced to increase the versa-
for stem cell labeling and multi-modal in vivo tracking. tility of SWNT-based imaging probes. Wang et al. for the first time
In conclusion, CNTs with appropriate modifications have been dem- reported the 125I could be used to track the biodistribution of SWNTs
onstrated to be useful contrast agents for both T1- and T2-weighted MR in animals [119]. An alternative method using 14C instead of 125I was
imaging. Particularly for T2-MR imaging, no additional treatment is also realized by the same group to track the long-term biodistribution
needed since the metallic impurities in the CNT samples could be uti- of MWNTs [120]. The modality of micro single photon emission com-
lized to offer the MR contrast. Moreover, different from the previously puterized tomography (micro-SPECT) was successfully obtained by la-
discussed optical-based imaging techniques, MR imaging enables beling CNTs with 111In [121]. Subsequently, McDevitt and coworkers
whole-body imaging without depth limit, and is thus a more clinical labeled SWNTs with 86Y for position emission tomography (PET) imag-
relevant imaging technique. Therefore, MR imaging with CNTs, if inte- ing [122]. In vivo tumor imaging in mouse models by radionuclide
grated with other abovementioned imaging techniques, may provide labeled SWNTs has also been reported by several groups [123,124]. In
novel opportunities in bio-imaging. 2007, Liu et al. used 64Cu to label PEGylated SWNTs with RGD peptide
1960 H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963

conjugation via a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic to the excitation laser would also significantly increase the Raman
acid (DOTA) chelator in vivo tumor-targeted PET imaging of U87MG signals of SWNTs on a per mass basis.
tumors using 64Cu/DOTA-labeled SWNT–PEG–RGD was realized owing Other imaging techniques using CNTs, including PA imaging, MR
to the specific binding between RGD peptide and integrin avβ3 over- imaging, and nuclear imaging, have also shown great potential in bio-
expressed on U87MG tumor cells as well as tumor vasculatures [123]. medical imaging. The integration of multiple imaging modalities could
In addition to RGD peptide, anti-CD20 antibody was also used to target extend the advantages of each single imaging modality and overcome
human Burkitt lymphoma by conjugating them with 111In labeled their inherent limitations. The real beauty of using CNTs as the imaging
SWNTs [124]. Besides using traditional chelation chemistry to obtain probe is thus their multi-functionalities. Unlike many other nano-
radiolabeled CNTs, radioisotopes could also be inserted into nanotubes materials used in bio-imaging, CNTs with highly enriched optical and
for radiolabeling [125]. Hong et al. reported that Na125I could be sealed magnetic properties are intrinsic multimodal imaging probes, without
inside the hollow structure of SWNTs, obtaining 125I-labeled SWNTs the need to engineer complicated nanostructures to afford multiple
useful for SPECT/CT imaging. Compared with the modalities mentioned functions. Moreover, the therapeutic functions of CNTs (e.g. drug & gene
above, radio-labeled SWNTs exhibit several advantages, such as no tis- delivery, photothermal therapy) [31,93,126] in combination with imaging
sue penetration depth limitation and high sensitivity. However, the would further make them excellent theranostic nano-platforms.
progress of using CNTs for nuclear imaging has become relatively slow Besides the limitations of each modality mentioned above, one
in the recent 2–3 years, without showing much significant advance to major challenge for the clinical application of CNTs is the concern of
our best knowledge. Nevertheless, the combination of nuclear imaging their potential long-term toxicity [127,128]. CNTs without appropriate
with other imaging techniques may still be of great interests in the surface functionalized have been found to be toxic in vivo, inducing a
development of CNT-based bio-imaging probes. wide range of toxicity to animals [129–131]. On the other hand, re-
searchers have also evidenced that CNTs with well design surface coat-
7. Prospects and challenges ings such as PEGylation exhibit no obvious in vitro and in vivo toxicity
[19,69,132–136], and may be gradually excreted from animals via
Different imaging modalities of SWNTs have their respective advan- renal and/or facial excretion pathways [137–140]. Several groups also
tages as well as limitations. The fluorescence imaging in the NIR-II claimed the possibility of biodegradation of CNTs under enzyme-
biological transparent window with SWNTs enables in vivo optical im- induced oxidization [141,142]. Nevertheless, it is expected that while
aging with deep tissue penetration and high spatial resolution, due to CNT-based biomedical imaging could indeed have the great potential
the low indigenous tissue scattering and the much reduced photon scat- in basic biomedical research as well as in vitro/ex vivo clinical diagnosis,
tering in the NIR-II region compared to that for fluorescence imaging there would still be significant challenges towards the use of those
in visible (400–700 nm) and traditional NIR-I region (700–900 nm). nanomaterials for real in vivo clinical applications.
However, the QY of SWNTs is still not high enough. Developing simple
synthesis and purification strategies to get SWNTs with pure chirality Acknowledgments
would be of great importance to obtain new SWNT-based NIR-II
imaging probes with bright and multi-colored fluorescence (Table 1). This work was partially supported by the National Natural Science
SWNTs have a large scattering cross-section and show strong reso- Foundation of China (51222203, 51002100, 51132006), the National
nance Raman scattering. Raman imaging by means of SWNT-probes “973” Program of China (2011CB911002, 2012CB932601), and a Project
has great sensitivity, and is resistant to photobleaching and quenching. Funded by the Priority Academic Program Development of Jiangsu
Relying on the SERS effect, noble metal coated SWNTs show further en- Higher Education Institutions.
hanced Raman signals, enabling Raman mapping with a much faster
speed. However, even with SERS enhancement, it still takes ~10 min References
to acquire a Raman image by conventional Raman mapping. The ad-
vances in instruments may be needed to make SWNT-based Raman im- [1] S. Iijima, Helical microtubules of graphitic carbon, Nature 354 (1991) 56–58.
[2] H.J. Dai, Carbon nanotubes: synthesis, integration, and properties, Acc. Chem. Res.
aging a viable tool in real biological research. Moreover, the use of 35 (2002) 1035–1044.
chirality-purified SWNTs with the majority of nanotubes in resonance [3] M.S. Dresselhaus, H. Dai, Carbon nanotubes: continued innovations and challenges,
MRS Bull. 29 (2004) 237–239.
[4] H. Ago, K. Petritsch, M.S. Shaffer, A.H. Windle, R.H. Friend, Composites of carbon
Table 1 nanotubes and conjugated polymers for photovoltaic devices, Adv. Mater. 11
(1999) 1281–1285.
Various modalities of imaging based on carbon nanotubes.
[5] M. Ouyang, J.L. Huang, C.M. Lieber, Fundamental electronic properties and applica-
Imaging modalities Examples References tions of single-walled carbon nanotubes, Acc. Chem. Res. 35 (2002) 1018–1025.
[6] N.S. Lee, D.S. Chung, I.T. Han, J.H. Kang, Y.S. Choi, H.Y. Kim, S.H. Park, Y.W. Jin, W.K.
NIR-II fluorescence imaging In vitro assay [56] Yi, M.J. Yun, J.E. Jung, C.J. Lee, J.H. You, S.H. Jo, C.G. Lee, J.M. Kim, Application of car-
In vivo whole animal imaging [20] bon nanotubes to field emission displays, Diam. Relat. Mater. 10 (2001) 265–270.
High frame rate fluorescent [26,28,43] [7] M.W. Rowell, M.A. Topinka, M.D. McGehee, H.-J. Prall, G. Dennler, N.S. Sariciftci, L.
video imaging combined with PCA Hu, G. Gruner, Organic solar cells with carbon nanotube network electrodes, Appl.
Chirality sorted SWNTs for in vivo [42] Phys. Lett. 88 (2006).
imaging [8] K. Besteman, J.O. Lee, F.G.M. Wiertz, H.A. Heering, C. Dekker, Enzyme-coated car-
Raman imaging In vitro cell imaging [27,69–71] bon nanotubes as single-molecule biosensors, Nano Lett. 3 (2003) 727–730.
[9] B.L. Allen, P.D. Kichambare, A. Star, Carbon nanotube field-effect-transistor-based
Multiplexed Raman imaging [22,78]
biosensors, Adv. Mater. 19 (2007) 1439–1451.
SERS enhanced Raman imaging [48,79–81]
[10] Z. Liu, K. Chen, C. Davis, S. Sherlock, Q. Cao, X. Chen, H. Dai, Drug delivery with car-
In vivo imaging [72,73] bon nanotubes for in vivo cancer treatment, Cancer Res. 68 (2008) 6652–6660.
Photoacoustic imaging Cell imaging by photoacoustic [95] [11] Z. Liu, X. Sun, N. Nakayama-Ratchford, H. Dai, Supramolecular chemistry on
Targeting in vivo PA imaging [19] water-soluble carbon nanotubes for drug loading and delivery, ACS Nano 1
GNTs for in vivo imaging and CTCs [21,96] (2007) 50–56.
Dye enhanced photoacoustic [49,97] [12] F. Zhou, D. Xing, Z. Ou, B. Wu, D.E. Resasco, W.R. Chen, Cancer photothermal ther-
imaging apy in the near-infrared region by using single-walled carbon nanotubes, J.
MR imaging T1 weighted MR imaging [107,108] Biomed. Opt. 14 (2009)(021009-021009).
T2 weighted MR imaging [32,109,110,114, [13] X. Liu, H. Tao, K. Yang, S. Zhang, S.-T. Lee, Z. Liu, Optimization of surface chemistry
115,118] on single-walled carbon nanotubes for in vivo photothermal ablation of tumors,
Nuclear imaging Biodistribution of SWNTs [119,120] Biomaterials 32 (2011) 144–151.
PET imaging [122,123] [14] G. Pastorin, W. Wu, S. Wieckowski, J.-P. Briand, K. Kostarelos, M. Prato, A. Bianco,
Double functionalisation of carbon nanotubes for multimodal drug delivery,
SPECT imaging [121,124,125]
Chem. Commun. (2006) 1182–1184.
H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963 1961

[15] R.P. Feazell, N. Nakayama-Ratchford, H. Dai, S.J. Lippard, Soluble single-walled [43] J.T. Robinson, G. Hong, Y. Liang, B. Zhang, O.K. Yaghi, H. Dai, In vivo fluorescence im-
carbon nanotubes as longboat delivery systems for platinum (IV) anticancer drug aging in the second near-infrared window with long circulating carbon nanotubes
design, J. Am. Chem. Soc. 129 (2007) 8438–8439. capable of ultrahigh tumor uptake, J. Am. Chem. Soc. 134 (2012) 10664–10669.
[16] J. Chen, S. Chen, X. Zhao, L.V. Kuznetsova, S.S. Wong, I. Ojima, Functionalized [44] T.K. Leeuw, R.M. Reith, R.A. Simonette, M.E. Harden, P. Cherukuri, D.A. Tsyboulski,
single-walled carbon nanotubes as rationally designed vehicles for tumor-targeted K.M. Beckingham, R.B. Weisman, Single-walled carbon nanotubes in the intact
drug delivery, J. Am. Chem. Soc. 130 (2008) 16778–16785. organism: Near-IR imaging and biocompatibility studies in Drosophila, Nano Lett.
[17] Z. Yinghuai, A.T. Peng, K. Carpenter, J.A. Maguire, N.S. Hosmane, M. Takagaki, 7 (2007) 2650–2654.
Substituted carborane-appended water-soluble single-wall carbon nanotubes: [45] P. Cherukuri, C.J. Gannon, T.K. Leeuw, H.K. Schmidt, R.E. Smalley, S.A. Curley, R.B.
new approach to boron neutron capture therapy drug delivery, J. Am. Chem. Soc. Weisman, Mammalian pharmacokinetics of carbon nanotubes using intrinsic
127 (2005) 9875–9880. near-infrared fluorescence, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 18882–18886.
[18] A.A. Bhirde, V. Patel, J. Gavard, G. Zhang, A.A. Sousa, A. Masedunskas, R.D. Leapman, [46] A.L. Antaris, J.T. Robinson, O.K. Yaghi, G. Hong, S. Diao, R. Luong, H. Dai, Ultra-low
R. Weigert, J.S. Gutkind, J.F. Rusling, Targeted killing of cancer cells in vivo and doses of chirality sorted (6,5) carbon nanotubes for simultaneous tumor imaging
in vitro with EGF-directed carbon nanotube-based drug delivery, ACS Nano 3 and photothermal therapy, ACS Nano 7 (2013) 3644–3652.
(2009) 307–316. [47] G. Hong, S.M. Tabakman, K. Welsher, H. Wang, X. Wang, H. Dai, Metal-enhanced
[19] L. Wu, X. Cai, K. Nelson, W. Xing, J. Xia, R. Zhang, A.J. Stacy, M. Luderer, G.M. Lanza, fluorescence of carbon nanotubes, J. Am. Chem. Soc. 132 (2010) 15920–15923.
L.V. Wang, A green synthesis of carbon nanoparticles from honey and their use in [48] X. Wang, C. Wang, L. Cheng, S.-T. Lee, Z. Liu, Noble metal coated single-walled
real-time photoacoustic imaging, Nano Res. 5 (2013) 312–325. carbon nanotubes for applications in surface enhanced Raman scattering imaging
[20] K. Welsher, Z. Liu, S.P. Sherlock, J.T. Robinson, Z. Chen, D. Daranciang, H. Dai, A and photothermal therapy, J. Am. Chem. Soc. 134 (2012) 7414–7422.
route to brightly fluorescent carbon nanotubes for near-infrared imaging in mice, [49] A.d.l. Zerda, Z. Liu, S. Bodapati, R. Teed, S. Vaithilingam, B.T. Khuri-Yakub, X. Chen,
Nat. Nanotechnol. 4 (2009) 773–780. H. Dai, S.S. Gambhir, Ultrahigh sensitivity carbon nanotube agents for photoacous-
[21] J.-W. Kim, E.I. Galanzha, E.V. Shashkov, H.-M. Moon, V.P. Zharov, Golden carbon tic molecular imaging in living mice, Nano Lett. 10 (2010) 2168–2172.
nanotubes as multimodal photoacoustic and photothermal high-contrast molecu- [50] X. He, J. Gao, S.S. Gambhir, Z. Cheng, Near-infrared fluorescent nanoprobes for
lar agents, Nat. Nanotechnol. 4 (2009) 688–694. cancer molecular imaging: status and challenges, Trends Mol. Med. 16 (2010)
[22] Z. Liu, X. Li, S.M. Tabakman, K. Jiang, S. Fan, H. Dai, Multiplexed multicolor Raman 574–583.
imaging of live cells with isotopically modified single walled carbon nanotubes, J. [51] B. Chance, Near-Infrared Images Using Continuous, Phase-Modulated, and Pulsed
Am. Chem. Soc. 130 (2008) 13540–13541. Light with Quantitation of Blood and Blood Oxygenation, in: R.R. Alfano (Ed.),
[23] C.L. Zavaleta, B.R. Smith, I. Walton, W. Doering, G. Davis, B. Shojaei, M.J. Natan, S.S. Advances in Optical Biopsy and Optical Mammography, 1998, pp. 29–45.
Gambhir, Multiplexed imaging of surface enhanced Raman scattering nanotags in [52] T. Ishizawa, N. Fukushima, J. Shibahara, K. Masuda, S. Tamura, T. Aoki, K. Hasegawa,
living mice using noninvasive Raman spectroscopy, Proc. Natl. Acad. Sci. 106 Y. Beck, M. Fukayama, N. Kokudo, Real-time identification of liver cancers by using
(2009) 13511–13516. indocyanine green fluorescent imaging, Cancer 115 (2009) 2491–2504.
[24] A.J. Beer, M. Schwaiger, Imaging of integrin αvβ3 expression, Cancer Metastasis [53] M. Sekijima, T. Tojimbara, S. Sato, M. Nakamura, T. Kawase, K. Kai, Y. Urashima, I.
Rev. 27 (2008) 631–644. Nakajima, S. Fuchinoue, S. Teraoka, An intraoperative fluorescent imaging system
[25] J.W.G. Wildoer, L.C. Venema, A.G. Rinzler, R.E. Smalley, C. Dekker, Electronic struc- in organ transplantation, Transplant. Proc. 36 (2004) 2188–2190.
ture of atomically resolved carbon nanotubes, Nature 391 (1998) 59–62. [54] S. Morimoto, In-vivo imaging of tumors with protease activated near-infrared
[26] K. Welsher, S.P. Sherlock, H. Dai, Deep-tissue anatomical imaging of mice using car- fluorescent probes, Tanpakushitsu kakusan koso, Protein, Nucleic Acid, Enzyme
bon nanotube fluorophores in the second near-infrared window, Proc. Natl. Acad. 52 (2007) 1774–1775.
Sci. 108 (2011) 8943–8948. [55] S. Kim, Y.T. Lim, E.G. Soltesz, A.M. De Grand, J. Lee, A. Nakayama, J.A. Parker, T.
[27] D.A. Heller, S. Baik, T.E. Eurell, M.S. Strano, Single‐walled carbon nanotube spec- Mihaljevic, R.G. Laurence, D.M. Dor, L.H. Cohn, M.G. Bawendi, J.V. Frangioni,
troscopy in live cells: towards long‐term labels and optical sensors, Adv. Mater. Near-infrared fluorescent type II quantum dots for sentinel lymph node mapping,
17 (2005) 2793–2799. Nat. Biotechnol. 22 (2004) 93–97.
[28] G. Hong, J.T. Robinson, J.P. Cooke, H. Dai, U. Raaz, L. Xie, N.F. Huang, J.C. Lee, Multi- [56] Y. Iizumi, T. Okazaki, Y. Ikehara, M. Ogura, S. Fukata, M. Yudasaka, Immunoassay
functional in vivo vascular imaging using near-infrared II fluorescence, Nat. Med. with single-walled carbon nanotubes as near-infrared fluorescent labels, ACS
18 (2012) 1841–1846. Appl. Mater. Interfaces (2013) 7665–7670.
[29] C.L. Zavaleta, B.R. Smith, I. Walton, W. Doering, G. Davis, B. Shojaei, M.J. Natan, S.S. [57] Y.T. Lim, S. Kim, A. Nakayama, N.E. Stott, M.G. Bawendi, J.V. Frangioni, Selection of
Gambhir, Multiplexed imaging of surface enhanced Raman scattering nanotags in quantum dot wavelengths for biomedical assays and imaging, Mol. Imaging 2
living mice using noninvasive Raman spectroscopy, Proc. Natl. Acad. Sci. U. S. A. (2003) 50–64.
106 (2009) 13511–13516. [58] S. Diao, G. Hong, J.T. Robinson, L. Jiao, A.L. Antaris, J.Z. Wu, C.L. Choi, H. Dai, Chirality
[30] A. De La Zerda, C. Zavaleta, S. Keren, S. Vaithilingam, S. Bodapati, Z. Liu, J. Levi, B.R. enriched (12, 1) and (11, 3) single-walled carbon nanotubes for biological imaging,
Smith, T.-J. Ma, O. Oralkan, Z. Cheng, X. Chen, H. Dai, B.T. Khuri-Yakub, S.S. J. Am. Chem. Soc. 134 (2012) 16971–16974.
Gambhir, Carbon nanotubes as photoacoustic molecular imaging agents in living [59] H. Yi, D. Ghosh, M.-H. Ham, J. Qi, P.W. Barone, M.S. Strano, A.M. Belcher, M13
mice, Nat. Nanotechnol. 3 (2008) 557–562. phage-functionalized single-walled carbon nanotubes as nanoprobes for second
[31] Z. Liu, S. Tabakman, K. Welsher, H. Dai, Carbon nanotubes in biology and medicine: near-infrared window fluorescence imaging of targeted tumors, Nano Lett. 12
in vitro and in vivo detection, imaging and drug delivery, Nano Res. 2 (2009) (2012) 1176–1183.
85–120. [60] H. Kuzmany, B. Burger, M. Fally, A.G. Rinzler, R.E. Smalley, Effect of dimensionality
[32] C. Wang, X. Ma, S. Ye, L. Cheng, K. Yang, L. Guo, C. Li, Y. Li, Z. Liu, Protamine func- in polymeric fullerenes and single-wall nanotubes, Physica B 244 (1998) 186–191.
tionalized single-walled carbon nanotubes for stem cell labeling and in vivo [61] A. Jorio, M. Pimenta, A. Souza Filho, R. Saito, G. Dresselhaus, M. Dresselhaus, Char-
Raman/magnetic resonance/photoacoustic triple-modal imaging, Adv. Funct. acterizing carbon nanotube samples with resonance Raman scattering, New J.
Mater. 22 (2012) 2363–2375. Phys. 5 (2003) 139.1–139.17.
[33] Z. Liu, W. Cai, L. He, N. Nakayama, K. Chen, X. Sun, X. Chen, H. Dai, In vivo [62] S. Ghosh, S.M. Bachilo, R.B. Weisman, Advanced sorting of single-walled carbon
biodistribution and highly efficient tumour targeting of carbon nanotubes in nanotubes by nonlinear density-gradient ultracentrifugation, Nat. Nanotechnol. 5
mice, Nat. Nanotechnol. 2 (2007) 47–52. (2010) 443–450.
[34] S.Y. Hong, G. Tobias, K.T. Al-Jamal, B. Ballesteros, H. Ali-Boucetta, S. Lozano-Perez, [63] X. Tu, S. Manohar, A. Jagota, M. Zheng, DNA sequence motifs for structure-specific
P.D. Nellist, R.B. Sim, C. Finucane, S.J. Mather, M.L.H. Green, K. Kostarelos, B.G. recognition and separation of carbon nanotubes, Nature 460 (2009) 250–253.
Davis, Filled and glycosylated carbon nanotubes for in vivo radioemitter localiza- [64] H. Liu, D. Nishide, T. Tanaka, H. Kataura, Large-scale single-chirality separation of
tion and imaging, Nat. Mater. 9 (2010) 485–490. single-wall carbon nanotubes by simple gel chromatography, Nat. Commun. 2
[35] H.-C. Wu, X. Chang, L. Liu, F. Zhao, Y. Zhao, Chemistry of carbon nanotubes in bio- (2011) 1–8.
medical applications, J. Mater. Chem. 20 (2010) 1036–1052. [65] Y. Miyata, K. Shiozawa, Y. Asada, Y. Ohno, R. Kitaura, T. Mizutani, H. Shinohara,
[36] K. Kostarelos, A. Bianco, M. Prato, Promises, facts and challenges for carbon nano- Length-sorted semiconducting carbon nanotubes for high-mobility thin film tran-
tubes in imaging and therapeutics, Nat. Nanotechnol. 4 (2009) 627–633. sistors, Nano Res. 4 (2011) 963–970.
[37] Z. Liu, K. Yang, S.-T. Lee, Single-walled carbon nanotubes in biomedical imaging, J. [66] K. Moshammer, F. Hennrich, M.M. Kappes, Selective suspension in aqueous sodium
Mater. Chem. 21 (2011) 586–598. dodecyl sulfate according to electronic structure type allows simple separation of
[38] H. Jin, D.A. Heller, M.S. Strano, Single-particle tracking of endocytosis and exocyto- metallic from semiconducting single-walled carbon nanotubes, Nano Res. 2
sis of single-walled carbon nanotubes in NIH-3T3 cells, Nano Lett. 8 (2008) (2009) 599–606.
1577–1585. [67] R. Martin, L. Falicov, Resonant Raman Scattering, Light scattering in Solids I, Springer,
[39] N.W.S. Kam, M. O'Connell, J.A. Wisdom, H.J. Dai, Carbon nanotubes as multifunc- 1983, pp. 79–145.
tional biological transporters and near-infrared agents for selective cancer cell [68] A. Rao, E. Richter, S. Bandow, B. Chase, P. Eklund, K. Williams, S. Fang, K.
destruction, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 11600–11605. Subbaswamy, M. Menon, A. Thess, Diameter-selective Raman scattering from vi-
[40] K. Welsher, Z. Liu, D. Daranciang, H. Dai, Selective probing and imaging of cells with brational modes in carbon nanotubes, Science 275 (1997) 187–191.
single walled carbon nanotubes as near-infrared fluorescent molecules, Nano Lett. [69] Z. Liu, C. Davis, W. Cai, L. He, X. Chen, H. Dai, Circulation and long-term fate of func-
8 (2008) 586–590. tionalized, biocompatible single-walled carbon nanotubes in mice probed by
[41] P. Cherukuri, S.M. Bachilo, S.H. Litovsky, R.B. Weisman, Near-infrared fluorescence Raman spectroscopy, Proc. Natl. Acad. Sci. 105 (2008) 1410–1415.
microscopy of single-walled carbon nanotubes in phagocytic cells, J. Am. Chem. [70] C. Lamprecht, N. Gierlinger, E. Heister, B. Unterauer, B. Plochberger, M.
Soc. 126 (2004) 15638–15639. Brameshuber, P. Hinterdorfer, S. Hild, A. Ebner, Mapping the intracellular distribu-
[42] S. Diao, G. Hong, J.T. Robinson, L. Jiao, A.L. Antaris, J.Z. Wu, C.L. Choi, H. Dai, Chirality tion of carbon nanotubes after targeted delivery to carcinoma cells using confocal
enriched (12,1) and (11,3) single-walled carbon nanotubes for biological imaging, Raman imaging as a label-free technique, J. Phys. Condens. Matter 24 (2012)
J. Am. Chem. Soc. 134 (2012) 16971–16974. 164206–164216.
1962 H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963

[71] D. Heller, J.W. Kang, N. Lue, R.R. Dasari, Measuring Uptake Dynamics Of Multiple, [101] A. Al Faraj, F. Fauvelle, N. Luciani, G. Lacroix, M. Levy, Y. Cremillieux, E. Canet-
Identifiable Carbon Nanotube Species Via High-Speed Confocal Raman Imaging Soulas, In vivo biodistribution and biological impact of injected carbon nanotubes
Of Live Cells, 223rd ECS Meeting (May 12–17, 2013), Ecs, 2013. using magnetic resonance techniques, Int. J. Nanomedicine 6 (2011) 351–361.
[72] C. Zavaleta, A. De La Zerda, Z. Liu, S. Keren, Z. Cheng, M. Schipper, X. Chen, H. Dai, S. [102] A. Cerpa, M. Koeber, D. Calle, V. Negri, J. Maria Gavira, A. Hernanz, F. Briones, S.
Gambhir, Noninvasive Raman spectroscopy in living mice for evaluation of tumor Cerdan, P. Ballesteros, Single-walled carbon nanotubes as anisotropic relaxation
targeting with carbon nanotubes, Nano Lett. 8 (2008) 2800–2805. probes for magnetic resonance imaging, MedChemComm 4 (2013) 669–672.
[73] B.R. Smith, C. Zavaleta, J. Rosenberg, R. Tong, J. Ramunas, Z. Liu, H. Dai, S.S. Gambhir, [103] B.-T. Doan, J. Seguin, M. Breton, R. Le Beherec, M. Bessodes, J.A. Rodriguez-Manzo, F.
High-resolution, serial intravital microscopic imaging of nanoparticle delivery and Banhart, J.-C. Beloeil, D. Scherman, C. Richard, Functionalized single-walled carbon
targeting in a small animal tumor model, Nano Today (2013) 126–137. nanotubes containing traces of iron as new negative MRI contrast agents for in vivo
[74] G.A. Wagnieres, W.M. Star, B.C. Wilson, In vivo fluorescence spectroscopy and imaging, Contrast Media Mol. Imaging 7 (2012) 153–159.
imaging for oncological applications, Photochem. Photobiol. 68 (1998) 603–632. [104] E.J. Rivera, R. Sethi, F. Qu, R. Krishnamurthy, R. Muthupillai, M. Alford, M.A.
[75] Y.C. Cao, R. Jin, C.A. Mirkin, Nanoparticles with Raman spectroscopic fingerprints Swanson, S.S. Eaton, G.R. Eaton, L.J. Wilson, Nitroxide radicals@US-tubes: new
for DNA and RNA detection, Science 297 (2002) 1536–1540. spin labels for biomedical applications, Adv. Funct. Mater. 22 (2012) 3691–3698.
[76] S. Keren, C. Zavaleta, Z. Cheng, A. de La Zerda, O. Gheysens, S. Gambhir, Noninvasive [105] B. Sitharaman, B.D. Jacobson, Y.Z. Wadghiri, H. Bryant, J. Frank, The magnetic,
molecular imaging of small living subjects using Raman spectroscopy, Proc. Natl. relaxometric, and optical properties of gadolinium-catalyzed single walled carbon
Acad. Sci. 105 (2008) 5844–5849. nanotubes, J. Appl. Phys. 113 (2013)(134308-134301-134308-134307).
[77] X. Li, X. Tu, S. Zaric, K. Welsher, W.S. Seo, W. Zhao, H. Dai, Selective synthesis com- [106] J.M. Tucker-Schwartz, T. Hong, D.C. Colvin, Y. Xu, M.C. Skala, Dual-modality
bined with chemical separation of single-walled carbon nanotubes for chirality se- photothermal optical coherence tomography and magnetic-resonance imaging of
lection, J. Am. Chem. Soc. 129 (2007) 15770–15771. carbon nanotubes, Opt. Lett. 37 (2012) 872–874.
[78] Z. Liu, S. Tabakman, S. Sherlock, X. Li, Z. Chen, K. Jiang, S. Fan, H. Dai, Multiplexed [107] J. Miyawaki, M. Yudasaka, H. Imai, H. Yorimitsu, H. Isobe, E. Nakamura, S. Iijima,
five-color molecular imaging of cancer cells and tumor tissues with carbon nano- Synthesis of ultrafine Gd2O3 nanoparticles inside single-wall carbon nanohorns,
tube Raman tags in the near-infrared, Nano Res. 3 (2010) 222–233. J. Phys. Chem. B 110 (2006) 5179–5181.
[79] D. Takagi, Y. Homma, H. Hibino, S. Suzuki, Y. Kobayashi, Single-walled carbon [108] C. Richard, B.-T. Doan, J.-C. Beloeil, M. Bessodes, É. Tóth, D. Scherman, Noncovalent
nanotube growth from highly activated metal nanoparticles, Nano Lett. 6 (2006) functionalization of carbon nanotubes with amphiphilic Gd3+ chelates: toward
2642–2645. powerful T1 and T2 MRI contrast agents, Nano Lett. 8 (2008) 232–236.
[80] D. Wang, Z.-C. Li, L. Chen, Templated synthesis of single-walled carbon nanotube [109] A. Al Faraj, K. Cieslar, G. Lacroix, S. Gaillard, E. Canet-Soulas, Y. Crémillieux, In vivo
and metal nanoparticle assemblies in solution, J. Am. Chem. Soc. 128 (2006) imaging of carbon nanotube biodistribution using magnetic resonance imaging,
15078–15079. Nano Lett. 9 (2009) 1023–1027.
[81] Y.C. Chen, R.J. Young, J.V. Macpherson, N.R. Wilson, Silver‐decorated carbon nano- [110] J.H. Choi, F.T. Nguyen, P.W. Barone, D.A. Heller, A.E. Moll, D. Patel, S.A. Boppart, M.S.
tube networks as SERS substrates, J. Raman Spectrosc. 42 (2011) 1255–1262. Strano, Multimodal biomedical imaging with asymmetric single-walled carbon
[82] H. Chu, J. Wang, L. Ding, D. Yuan, Y. Zhang, J. Liu, Y. Li, Decoration of gold nanopar- nanotube/iron oxide nanoparticle complexes, Nano Lett. 7 (2007) 861–867.
ticles on surface-grown single-walled carbon nanotubes for detection of every [111] O. Vittorio, S. Duce, V. Raffa, A. Cuschieri, Imaging and Biomedical Application of
nanotube by surface-enhanced Raman spectroscopy, J. Am. Chem. Soc. 131 Magnetic Carbon Nanotubes, 2011. 189–210.
(2009) 14310–14316. [112] Y.A. Mackeyev, J.W. Marks, M.G. Rosenblum, L.J. Wilson, Stable containment of ra-
[83] Z. Chen, S.M. Tabakman, A.P. Goodwin, M.G. Kattah, D. Daranciang, X. Wang, G. dionuclides on the nanoscale by cut single-wall carbon nanotubes, J. Phys. Chem. B
Zhang, X. Li, Z. Liu, P.J. Utz, Protein microarrays with carbon nanotubes as multicol- 109 (2005) 5482–5484.
or Raman labels, Nat. Biotechnol. 26 (2008) 1285–1292. [113] M. Monthioux, Filling single-wall carbon nanotubes, Carbon 40 (2002) 1809–1823.
[84] L. Beqa, Z. Fan, A.K. Singh, D. Senapati, P.C. Ray, Gold nano-popcorn attached [114] H. Wu, G. Liu, X. Wang, J. Zhang, Y. Chen, J. Shi, H. Yang, H. Hu, S. Yang,
SWCNT hybrid nanomaterial for targeted diagnosis and photothermal therapy of Solvothermal synthesis of cobalt ferrite nanoparticles loaded on multiwalled car-
human breast cancer cells, ACS Appl. Mater. Interfaces 3 (2011) 3316–3324. bon nanotubes for magnetic resonance imaging and drug delivery, Acta Biomater.
[85] L. Beqa, A.K. Singh, Z. Fan, D. Senapati, P.C. Ray, Chemically attached gold nanopar- 7 (2011) 3496–3504.
ticle–carbon nanotube hybrids for highly sensitive SERS substrate, Chem. Phys. [115] J.S. Ananta, M.L. Matson, A.M. Tang, T. Mandal, S. Lin, K. Wong, S.T. Wong, L.J.
Lett. 512 (2011) 237–242. Wilson, Single-walled carbon nanotube materials as T 2-weighted MRI contrast
[86] X. Wang, Y. Pang, G. Ku, X. Xie, G. Stoica, L.V. Wang, Noninvasive laser-induced agents, J. Phys. Chem. C 113 (2009) 19369–19372.
photoacoustic tomography for structural and functional in vivo imaging of the [116] J.M. Gimble, A.J. Katz, B.A. Bunnell, Adipose-derived stem cells for regenerative
brain, Nat. Biotechnol. 21 (2003) 803–806. medicine, Circ. Res. 100 (2007) 1249–1260.
[87] G. Ku, L.V. Wang, Deeply penetrating photoacoustic tomography in biological [117] A.I. Caplan, Adult mesenchymal stem cells for tissue engineering versus regenera-
tissues enhanced with an optical contrast agent, Opt. Lett. 30 (2005) 507–509. tive medicine, J. Cell. Physiol. 213 (2007) 341–347.
[88] C. Hoelen, F. De Mul, R. Pongers, A. Dekker, Three-dimensional photoacoustic [118] O. Vittorio, S.L. Duce, A. Pietrabissa, A. Cuschieri, Multiwall carbon nanotubes as
imaging of blood vessels in tissue, Opt. Lett. 23 (1998) 648–650. MRI contrast agents for tracking stem cells, Nanotechnology 22 (2011) 095706.
[89] K. Yang, L. Hu, X. Ma, S. Ye, L. Cheng, X. Shi, C. Li, Y. Li, Z. Liu, Multimodal imaging [119] H. Wang, J. Wang, X. Deng, H. Sun, Z. Shi, Z. Gu, Y. Liu, Y. Zhaoc, Biodistribution of
guided photothermal therapy using functionalized graphene nanosheets anchored carbon single-wall carbon nanotubes in mice, J. Nanosci. Nanotechnol. 4 (2004)
with magnetic nanoparticles, Adv. Mater. 24 (2012) 1868–1872. 1019–1024.
[90] G. Ku, M. Zhou, S. Song, Q. Huang, J. Hazle, C. Li, Copper sulfide nanoparticles as a [120] X. Deng, S. Yang, H. Nie, H. Wang, Y. Liu, A generally adoptable radiotracing method
new class of photoacoustic contrast agent for deep tissue imaging at 1064 nm, for tracking carbon nanotubes in animals, Nanotechnology 19 (2008) 075101.
ACS Nano 6 (2012) 7489–7496. [121] L. Lacerda, A. Soundararajan, R. Singh, G. Pastorin, K.T. Al‐Jamal, J. Turton, P.
[91] A. Agarwal, S. Huang, M. ODonnell, K. Day, M. Day, N. Kotov, S. Ashkenazi, Targeted Frederik, M.A. Herrero, S. Li, A. Bao, Dynamic imaging of functionalized multi‐
gold nanorod contrast agent for prostate cancer detection by photoacoustic imag- walled carbon nanotube systemic circulation and urinary excretion, Adv. Mater.
ing, J. Appl. Phys. 102 (2007) 064701–064704. 20 (2008) 225–230.
[92] J.T. Robinson, K. Welsher, S.M. Tabakman, S.P. Sherlock, H. Wang, R. Luong, H. Dai, [122] M.R. McDevitt, D. Chattopadhyay, J.S. Jaggi, R.D. Finn, P.B. Zanzonico, C. Villa, D. Rey,
High performance in vivo near-IR (N1 μm) imaging and photothermal cancer ther- J. Mendenhall, C.A. Batt, J.T. Njardarson, PET imaging of soluble yttrium-86-labeled
apy with carbon nanotubes, Nano Res. 3 (2010) 779–793. carbon nanotubes in mice, PLoS One 2 (2007) e907.
[93] X. Liu, H. Tao, K. Yang, S. Zhang, S.-T. Lee, Z. Liu, Optimization of surface chemistry [123] Z. Liu, W. Cai, L. He, N. Nakayama, K. Chen, X. Sun, X. Chen, H. Dai, In vivo
on single-walled carbon nanotubes for in vivo photothermal ablation of tumors, biodistribution and highly efficient tumour targeting of carbon nanotubes in
Biomaterials 32 (2011) 144–151. mice, Nat. Nanotechnol. 2 (2006) 47–52.
[94] H.K. Moon, S.H. Lee, H.C. Choi, In vivo near-infrared mediated tumor destruction by [124] M.R. McDevitt, D. Chattopadhyay, B.J. Kappel, J.S. Jaggi, S.R. Schiffman, C. Antczak,
photothermal effect of carbon nanotubes, ACS Nano 3 (2009) 3707–3713. J.T. Njardarson, R. Brentjens, D.A. Scheinberg, Tumor targeting with antibody-
[95] P.K. Avti, S. Hu, C. Favazza, A.G. Mikos, J.A. Jansen, K.R. Shroyer, L.V. Wang, B. functionalized, radiolabeled carbon nanotubes, J. Nucl. Med. 48 (2007) 1180–1189.
Sitharaman, Detection, mapping, and quantification of single walled carbon nano- [125] S.Y. Hong, G. Tobias, K.T. Al-Jamal, B. Ballesteros, H. Ali-Boucetta, S. Lozano-Perez,
tubes in histological specimens with photoacoustic microscopy, PloS One 7 (2012) P.D. Nellist, R.B. Sim, C. Finucane, S.J. Mather, Filled and glycosylated carbon nano-
e35064. tubes for in vivo radioemitter localization and imaging, Nat. Mater. 9 (2010)
[96] E.I. Galanzha, E.V. Shashkov, T. Kelly, J.-W. Kim, L. Yang, V.P. Zharov, In vivo mag- 485–490.
netic enrichment and multiplex photoacoustic detection of circulating tumour [126] A. Bianco, K. Kostarelos, M. Prato, Applications of carbon nanotubes in drug delivery,
cells, Nat. Nanotechnol. 4 (2009) 855–860. Curr. Opin. Chem. Biol. 9 (2005) 674–679.
[97] A. de la Zerda, S. Bodapati, R. Teed, S.n.Y. May, S.M. Tabakman, Z. Liu, B.T. [127] K. Yang, Z. Liu, In vivo biodistribution, pharmacokinetics, and toxicology of carbon
Khuri-Yakub, X. Chen, H. Dai, S.S. Gambhir, Family of enhanced photoacoustic im- nanotubes, Curr. Drug Metab. 13 (2012) 1057–1067.
aging agents for high-sensitivity and multiplexing studies in living mice, ACS Nano [128] L. Lacerda, A. Bianco, M. Prato, K. Kostarelos, Carbon nanotubes as nanomedicines:
6 (2012) 4694–4701. from toxicology to pharmacology, Adv. Drug Deliv. Rev. 58 (2006) 1460–1470.
[98] A.E. Merbach, É. Tóth, The Chemistry of Contrast Agents in Medical Magnetic [129] D.B. Warheit, B. Laurence, K.L. Reed, D. Roach, G. Reynolds, T. Webb, Comparative
Resonance Imaging, Wiley Chichester (W. Sx.) etc., 2001 pulmonary toxicity assessment of single-wall carbon nanotubes in rats, Toxicol.
[99] B. Sitharaman, K.R. Kissell, K.B. Hartman, L.A. Tran, A. Baikalov, I. Rusakova, Y. Sun, Sci. 77 (2004) 117–125.
H.A. Khant, S.J. Ludtke, W. Chiu, Superparamagnetic gadonanotubes are high- [130] C.-W. Lam, J.T. James, R. McCluskey, R.L. Hunter, Pulmonary toxicity of single-wall
performance MRI contrast agents, Chem. Commun. (2005) 3915–3917. carbon nanotubes in mice 7 and 90 days after intratracheal instillation, Toxicol. Sci.
[100] A. Al Faraj, K. Cieslar, G. Lacroix, S. Gaillard, E. Canot-Soulas, Y. Cremillieux, In vivo 77 (2004) 126–134.
imaging of carbon nanotube biodistribution using magnetic resonance imaging, [131] C.A. Poland, R. Duffin, I. Kinloch, A. Maynard, W.A. Wallace, A. Seaton, V. Stone, S.
Nano Lett. 9 (2009) 1023–1027. Brown, W. MacNee, K. Donaldson, Carbon nanotubes introduced into the
H. Gong et al. / Advanced Drug Delivery Reviews 65 (2013) 1951–1963 1963

abdominal cavity of mice show asbestos-like pathogenicity in a pilot study, Nat. administered carbon nanotube radiotracers, Proc. Natl. Acad. Sci. U. S. A. 103
Nanotechnol. 3 (2008) 423–428. (2006) 3357–3362.
[132] H. Dumortier, S. Lacotte, G. Pastorin, R. Marega, W. Wu, D. Bonifazi, J.-P. Briand, M. [138] Z. Liu, C. Davis, W. Cai, L. He, X. Chen, H. Dai, Circulation and long-term fate of func-
Prato, S. Muller, A. Bianco, Functionalized carbon nanotubes are non-cytotoxic and tionalized, biocompatible single-walled carbon nanotubes in mice probed by
preserve the functionality of primary immune cells, Nano Lett. 6 (2006) 1522–1528. Raman spectroscopy, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 1410–1415.
[133] S.-T. Yang, X. Wang, G. Jia, Y. Gu, T. Wang, H. Nie, C. Ge, H. Wang, Y. Liu, Long-term [139] L. Lacerda, A. Soundararajan, R. Singh, G. Pastorin, K.T. Al-Jamal, J. Turton, P.
accumulation and low toxicity of single-walled carbon nanotubes in intravenously Frederik, M.A. Herrero, S.L.A. Bao, D. Emfietzoglou, S. Mather, W.T. Phillips, M.
exposed mice, Toxicol. Lett. 181 (2008) 182–189. Prato, A. Bianco, B. Goins, K. Kostarelos, Dynamic Imaging of functionalized
[134] I. Hamad, A. Hunter, J. Szebeni, S.M. Moghimi, Poly (ethylene glycol) s generate multi-walled carbon nanotube systemic circulation and urinary excretion, Adv.
complement activation products in human serum through increased alternative Mater. 20 (2008) 225–230.
pathway turnover and a MASP-2-dependent process, Mol. Immunol. 46 (2008) [140] L. Lacerda, M.A. Herrero, K. Venner, A. Bianco, M. Prato, K. Kostarelos, Carbon-
225–232. nanotube shape and individualization critical for renal excretion, Small 4 (2008)
[135] J. Kolosnjaj-Tabi, K.B. Hartman, S. Boudjemaa, J.S. Ananta, G. Morgant, H. Szwarc, 1130–1132.
L.J. Wilson, F. Moussa, In vivo behavior of large doses of ultrashort and [141] B.L. Allen, P.D. Kichambare, P. Gou, I.I. Vlasova, A.A. Kapralov, N. Konduru, V.E.
full-length single-walled carbon nanotubes after oral and intraperitoneal adminis- Kagan, A. Star, Biodegradation of single-walled carbon nanotubes through enzy-
tration to Swiss mice, ACS Nano 4 (2010) 1481–1492. matic catalysis, Nano Lett. 8 (2008) 3899–3903.
[136] X. Chen, U.C. Tam, J.L. Czlapinski, G.S. Lee, D. Rabuka, A. Zettl, C.R. Bertozzi, Interfac- [142] V.E. Kagan, N.V. Konduru, W. Feng, B.L. Allen, J. Conroy, Y. Volkov, I.I. Vlasova,
ing carbon nanotubes with living cells, J. Am. Chem. Soc. 128 (2006) 6292–6293. N.A. Belikova, N. Yanamala, A. Kapralov, Carbon nanotubes degraded by
[137] R. Singh, D. Pantarotto, L. Lacerda, G. Pastorin, C. Klumpp, M. Prato, A. Bianco, K. neutrophil myeloperoxidase induce less pulmonary inflammation, Nat. Nanotechnol. 5
Kostarelos, Tissue biodistribution and blood clearance rates of intravenously (2010) 354–359.

You might also like