You are on page 1of 13

Pharmacology & Therapeutics 121 (2009) 192204

Contents lists available at ScienceDirect

Pharmacology & Therapeutics


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / p h a r m t h e r a

Associate editor: J.S. Fedan

Mechanisms of pulmonary toxicity and medical applications of carbon nanotubes:


Two faces of Janus?
A.A. Shvedova a,b,, E.R. Kisin a, D. Porter a,b, P. Schulte c, V.E. Kagan d, B. Fadeel e, V. Castranova a
a

Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, United States
Education and Information Division, National Institute for Occupational Safety and Health, Cincinnati, OH, United States
d
Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
e
Division of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
b
c

a r t i c l e

i n f o

Keywords:
Carbon nanotubes
Nanotoxicology
Cellular interactions
Pulmonary toxicity
Biomedical applications.

a b s t r a c t
Nanotechnology is an emerging science involving manipulation of materials at the nanometer scale. There
are several exciting prospects for the application of engineered nanomaterials in medicine. However,
concerns over adverse and unanticipated effects on human health have also been raised. In fact, the same
properties that make engineered nanomaterials attractive from a technological and biomedical perspective
could also make these novel materials harmful to human health and the environment. Carbon nanotubes are
cylinders of one or several coaxial graphite layer(s) with a diameter in the order of nanometers, and serve as
an instructive example of the Janus-like properties of nanomaterials. Numerous in vitro and in vivo studies
have shown that carbon nanotubes and/or associated contaminants or catalytic materials that arise during
the production process may induce oxidative stress and prominent pulmonary inammation. Recent studies
also suggest some similarities between the pathogenic properties of multi-walled carbon nanotubes and
those of asbestos bers. On the other hand, carbon nanotubes can be readily functionalized and several
studies on the use of carbon nanotubes as versatile excipients for drug delivery and imaging of disease
processes have been reported, suggesting that carbon nanotubes may have a place in the armamentarium for
treatment and monitoring of cancer, infection, and other disease conditions. Nanomedicine is an emerging
eld that holds great promise; however, close attention to safety issues is required to ensure that the
opportunities that carbon nanotubes and other engineered nanoparticles offer can be translated into feasible
and safe constructs for the treatment of human disease.
Published by Elsevier Inc.

Contents
1.
2.
3.
4.
5.
6.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . .
In vitro evaluation of the bioactivity of carbon nanotubes . . . . . . .
In vivo evaluation of the bioactivity of single-walled carbon nanotubes . . .
In vivo evaluation of the bioactivity of multi-walled carbon nanotubes .
Genotoxicity and mutagenicity of carbon nanotubes . . . . . . . . .
Perspectives on medical applications of carbon nanotubes . . . . . .

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

193
193
195
197
198
199

Abbreviations: CNT, carbon nanotubes; CoMoCAT, cobaltmolybdenum catalyst process; DSWCNT, dispersed single-walled carbon nanotubes; DTPA, diethylenetriaminepentaacetic; EPA, Environmental Protection Agency; HARN, high aspect ratio nanoparticles; HiPco, high-pressure carbon monoxide process; IT, intratracheal; LM, listeria monocytogenes;
MN, micronucleus; MTT, [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide]; MWCNT, multi-walled carbon nanotubes; NADPH, nicotinamide adenine dinucleotide
phosphate; OSHA, Occupational Safety and Health Administration; PEL, permissible exposure level; PMNs, polymorphonuclear neutrophils; PS, phosphatidylserine; ROS, reactive
oxygen species; SOD, superoxide dismutase; SWCNT, single-walled carbon nanotubes; TSCA, Toxic Substances Control Act.
Disclaimer: The ndings and conclusions in this report are those of the authors and do not necessarily represent the views of the National Institute for Occupational Safety and
Health.
Corresponding author. Health Effects Laboratory Division, NIOSH, Morgantown, WV 26505, United States. Tel.: 304 285 6177; fax: 304 285 5938.
E-mail address: ats1@cdc.gov (A.A. Shvedova).
0163-7258/$ see front matter. Published by Elsevier Inc.
doi:10.1016/j.pharmthera.2008.10.009

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

7.
Regulatory issues related
8.
Concluding remarks . .
Acknowledgments . . . . . . .
References . . . . . . . . . . .

to carbon
. . . . .
. . . . .
. . . . .

nanotubes
. . . . .
. . . . .
. . . . .

.
.
.
.

.
.
.
.

1. Introduction
Nanotechnology presents many opportunities and benets for new
materials with signicantly improved properties as well as revolutionary applications in the elds of energy, environment, medicine,
etc. The industrial production and use of nanoparticles will be the
driving force for the emerging new materials industry of the 21st
century. In terms of economics, analysts have estimated that the
worldwide market for nanomaterials will be 7001000 billion Euros
in 2011 (www.nanosafe2008.org). However, the potential impact of
these new materials on human health and the environment is viewed
with apprehension. Carbon nanotubes (CNT) are among newly
developed products and are currently of interest for a variety of
applications in electronics, reinforced rods, micro-fabricating conjugated polymer activators, biosensors, enhanced electron/scanning
microscopy imaging techniques, etc. Engineered nanoparticles are
already in consumer products, such as sun screens, cosmetics and
toiletry products, which are used daily by millions. The market for
these materials is estimated to grow to over eight billion dollars in the
next decade. Introduction of novel materials into industry requires
evaluation of safety and an understanding of the impact of CNT on the
environment, biological species and human health.
Discovered in 1991 by Iijima (1991), CNT represent allotropes of
carbon with a nanostructure that can have a length-to-diameter
ratio greater than 1,000,000. CNT can be viewed as a sheet of
graphite (a hexagonal lattice of carbon) rolled into a cylinder. Due to
their unique physical and chemical properties, these structures have
sparked much interest recently with a large amount of research
dedicated to their novel applications. A very broad range of physical
properties of CNTaffecting their electronic, thermal, and structural
characteristicsis dened by their diameter, length, and chirality or
twist. In addition to single-walled carbon nanotubes (SWCNT) with a
single cylindrical carbon wall, multi-walled carbon nanotubes
(MWCNT) have multiple wallscylinders nested within other
cylinders.
There are several major techniques of CNT synthesis (Awasthi et al.,
2005; Kingston & Simard, 2006). The arc-evaporation method, which
produces good quality nanotubes, involves passing a current of about
50 A between two graphite electrodes in an atmosphere of helium in
the presence of metal catalysts (Co or Ni). The second method is
chemical vapor deposition, where nanotubes are formed by the
decomposition of a carbon-containing gas with use of nano-sized
catalytic particles usually Fe, Co, Yt or Ni. The advantage of catalytic
synthesis over arc-evaporation is the ability to scale-up for volume
production. The most commonly used production technologies are the
HiPco process, and the CoMoCAT process. The HiPco production
technology is a gas-phase homogeneous process that employs a
oating catalyst approach, whereby the growth catalyst is formed in
situ during the production. CNT are produced from the disproportionation of carbon monoxide over catalytic iron nanoparticles. In the
HiPco process, a multi-step purication approach, involving oxidation,
acid treatment and ltration, is commonly used to remove amorphous
carbon and residual iron impurities from the nal carbon nanotube
product. The CoMoCAT production technology is a heterogeneous
process involving growth on supported catalysts. CNT are produced by
the catalytic decomposition of carbon monoxide on silica supported,
CoMo bimetallic catalyst particles. The silica supports, residual cobalt
and molybdenum particles, and amorphous carbon are removed from
the nal CNT product by silica leaching, froth otation, acid treatment
and ltration purication processes. The third important method for

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

193

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

.
.
.
.

200
201
202
202

making CNT, laser ablation, involves employment of a powerful laser


to vaporize metal (Co and Ni)-graphite target. Of the three major types
of technological processes described, chemical vapor deposition is the
most prominent one that is currently used for CNT production; it is
providing not only higher volume of products but also CNT materials
containing less residual metal catalysts.
Rapid growth and escalation in manufacturing of nanomaterials
necessitate establishing control and withstanding possible adverse
health effects that may take place throughout the production or use.
The unique characteristics of nanomaterials (e.g. high surface area and
reactivity, ability to bind and deliver other molecules to target objects)
that are essential for the successful applications, might lead to
negative health impacts (Kagan et al., 2005; Fadeel et al., 2007). The
rapid progress in the development and use of nanomaterials is not yet
matched by adequate toxicological investigations. Experimental
animal studies suggest that the increased concentration of nanoparticles and higher reactive surface area per unit mass, along with
unique chemistry and functionality, is important in the acute
inammatory and chronic response in lung (Fadeel et al., 2007). The
exact relationship between the physicalchemical properties of
nanoparticles, their cell-to-cell interactions, reactivity, and biological/systemic consequences cannot be predicted. It is important to
know such inter-relationships beforehand to employ the benets of
nanotechnology without the hazardous consequences.
2. In vitro evaluation of the bioactivity of carbon nanotubes
Although industrial scale production and commercialization of
carbon nanotubes into various products is in the early stages, a
substantial number of studies have been published evaluating the
effects of in vitro exposure to SWCNT or MWCNT. This section will
review the results of these studies, highlight critical issues concerning
the conduct and evaluation of in vitro studies, and correlate in vitro
results with those from animal model studies where appropriate.
2.1. Cellular uptake of CNT
A number of recent studies addressed the issue of SWCNT uptake
by different cells. While the results seem to be controversial, it seems
apparent that the presence or absence of specialized signals
determines the recognition and subsequent interactions of SWCNT
with cells. Overall, pristine SWCNT carrying no recognizable signals
are poorly taken-up whereas SWCNT modied chemically (e.g.,
oxidatively modied during purication and storage) or by adsorbed
macromolecules (e.g., proteins, lipids) are more readily recognized
and engulfed by cells.
Several in vitro studies support the concept that SWCNT are not
readily taken up by lung cells. Davoren et al. (2007) reported no
measurable uptake of SWCNT in A549 cells (a human alveolar type II
cancer cell line). Likewise, Herzog et al. (2007) reported no uptake of
SWCNT in either A549 cells or BEAS-2B cells (a human bronchial
epithelial cell line). Lastly, no evidence of uptake of SWCNT was
reported after electron microscopic evaluation of exposed RAW 264.7
cells (a mouse peritoneal macrophage cell line) (Shvedova et al.,
2005). Mercer et al. (2008) investigated the deposition and fate of
gold-labeled SWCNT after aspiration in a mouse model. Results
indicate that disperse SWCNT structures were not avidly recognized
and engulfed by alveolar macrophages. Rather, the vast majority of
SWCNT escaped phagocytosis and rapidly migrated into the interstitium of the alveolar septa.

194

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

In contrast, signicant uptake of SWCNT has been reported in RAW


264.7 cells or NR8383 cells (a rat alveolar macrophage cell line) (Dutta
et al., 2007; Pulskump et al., 2007). Likewise, Jia et al. (2005) reported
uptake of SWCNT and MWCNT by primary guinea pig alveolar
macrophages after in vitro exposure. Interestingly, in studies which
reported cellular uptake, CNT were suspended in media containing
high serum concentrations (1015% fetal bovine or calf serum). Dutta
et al. (2007) reported that when suspended in a high serum medium
CNT avidly adsorb albumin. Upon binding to the surface of CNT,
albumin is structurally altered and is recognized by scavenger
receptors. Therefore, the reported uptake of CNT appears to be due
to an albumin coating rather than avid cellular recognition of the CNT
themselves. In these studies, the 1015% serum media used to
improve dispersion of the CNT affected the bioactivity of the CNT. To
avoid this problem, Porter et al. (2008) developed a dispersion
medium which signicantly decreased agglomeration of MWCNT but
did not affect particle bioactivity. This dispersion medium contained
low concentrations of disaturated phosphatidylcholine (0.01 mg/ml)
and serum albumin (0.6 mg/ml), which were sufcient to hinder van
der Waals forces but not to mask the surface of CNT. Therefore, this
dispersion medium may be useful in in vitro studies of the bioactivity
of CNT. In contrast, coating of SWCNT with a phospholipid eat-me
signal, phosphatidylserine, made them recognizable in vitro by
different phagocytic cells, including murine RAW264.7 macrophages,
primary monocyte-derived human macrophages and dendritic cells,
and microglia from rat brain (Fig. 1) (Konduru et al., submitted for
publication). Recognition and uptake were suppressed by the PSbinding protein, annexin V. Loading of PS-coated SWCNT with proapoptotic cargo (cytochrome c) allowed for the targeted killing of
RAW264.7 macrophages. In line with this, in vivo aspiration of PScoated SWCNT stimulated their uptake by lung alveolar macrophages
in mice.

Several studies using non-dye assays of viability have reported


cytotoxicity for CNT. SWCNT have been reported to decrease cell
number with RAW 264.7 macrophages (Dutta et al., 2007), decrease
colony size with A549 or BEAS-2B cells (Herzog et al., 2007), and
decrease of cell proliferation with mesothelial cells (Wick et al., 2006,
2007). MWCNT have also been reported to be cytotoxic to lung tumor
cells (Magrez et al., 2006). However, an increase in the agglomeration
state of CNT and/or acid oxidation of the CNT surface appear to be
related to this reported cytotoxicity (Wick et al., 2006, 2007; Davoren
et al., 2007; Dutta et al., 2007; Herzog et al., 2007). Therefore, it may be
prudent to conduct in vitro cytotoxicity testing of nanoparticles using
well dispersed suspensions (Sager et al., 2007; Porter et al., 2008).
The degree of metal catalyst contamination of the CNT sample
also appears to be a factor in the magnitude of in vitro cytotoxicity.
Shvedova et al. (2007b) reported that SWCNT synthesized by the
HiPco or laser ablation and having a content of 30% iron or 20%
nickel, respectively, generated hydroxyl radicals and caused a
signicant decrease in viability and decline in glutathione levels in
BEAS-2B cells. Iron-contaminated SWCNT were also reported in
other studies to exhibit cytotoxicity in A549 or BEAS-2B cells
(Herzog et al., 2007), BEAS-2B cells (Shvedova et al., 2004), and RAW
264.7 cells (Kagan et al., 2006). However, SWCNT samples with low
iron contamination were found to be signicantly less toxic (Kagan
et al., 2006; Herzog et al., 2007), while treatment of raw SWCNT
with a chelator to remove contaminating iron has been shown to
signicantly decrease both oxidant generation and cytotoxicity
(Shvedova et al., 2004).
In conclusion, the cytotoxicity studies for CNT may at rst glance
appear to be in conict. However, if one considers interference with

2.2. Cellular toxicity


Recently, Murr et al. (2005) reported that in vitro exposure to
SWCNT or MWCNT caused asbestos-like cytotoxicity in RAW 264.7
cells determined using the MTT assay. Similar results using the MTT
viability assay were reported for RAW 264.7 cells (Soto et al., 2005), for
RAW 264.7, A549, and THB-1 human macrophages (Soto et al., 2007),
and for guinea pig alveolar macrophages (Jia et al., 2005). Using
alamar blue, neutral red and MTT viability assays, Davoren et al.
(2007) reported a substantially lower level of cytotoxicity for SWCNT
in A549 cells. In contrast, two other studies reported low cytotoxicity
for raw SWCNT (high iron contamination), puried SWCNT (low iron),
and puried MWCNT (low iron) in A549 cells and NR8383 rat alveolar
macrophages (Worle-Knirsch et al., 2006; Pulskump et al., 2007).
Although these authors reported cytotoxicity using the MTT assay,
no cytotoxicity was demonstrated using lactate dehydrogenase,
propidium iodide, WST-1 (4-[3-(4-iodophenyl)-2-(4-nitrophenyl)2H-5-tetrazolio]-1,3-benzene disulfonate), apoptotic, or mitochondrial membrane potential assays for viability. Likewise, Shvedova et al.
(2005) reported that in vitro exposure of RAW 264.7 cells to puried
SWCNT did not cause apoptosis. Furthermore, Yacobi et al. (2007)
reported only a transient decrease in alveolar epithelial cell monolayers after exposure to SWCNT. In contrast, ambient ultrane particulate matter and quantum dots caused a large and prolonged effect.
Recently, several investigators have cautioned that CNT can
interfere with several dye-base viability assays (Monteiro-Riviere &
Inman, 2006; Worle-Knirsch et al., 2006; Davoren et al., 2007;
Pulskump et al., 2007). In addition to physical interference due to light
absorbance and scattering, Casey et al. (2007) has reported spectroscopic evidence that SWCNT interacts with various dyes commonly
used to assess cytotoxicity. Therefore, the studies using the MTT dyebased viability assay to report high cytotoxicity of CNT are now in
question.

Fig. 1. Representative transmission electron (A) and scanning electron (B) micrographs
of RAW264.7 macrophages with engulfed PS-coated SWCNT. RAW264.7 macrophages
(0.3 106 cells/ml) were incubated for 2 h with PS-coated SWCNT. Arrows indicate
SWCNT.

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

dye-based viability assays, agglomeration issues related to the method


of dispersion, and oxidant stress due to metal contamination of CNT,
the data available to date favor the conclusion that well-dispersed,
puried CNT exhibit relatively low cytotoxicity in vitro.

195

indicating that CNT can reach the pleural space is currently available.
Therefore, such studies are warranted.
3. In vivo evaluation of the
bioactivity of single-walled carbon nanotubes

2.3. Stimulation of inammatory factors and oxidants


Shvedova et al. (2005) reported that aspiration of SWCNT resulted
in a rapid but transient inammatory reaction in a mouse model. In
vitro studies support the conclusion that puried CNT are not potent
inducers of oxidant species or inammatory mediators. Kagan et al.
(2006) reported that SWCNT did not stimulate intracellular ROS or
nitric oxide (NO) production in RAW 264.7 cells. Dutta et al. (2007)
have shown that SWCNT failed to stimulate cyclooxygenase-2 by RAW
264.7 cells. Shvedova et al. (2005) reported that SWCNT failed to
stimulate intracellular ROS, NO production, or the secretion of TNF-
and IL-1 in RAW 264.7 cells. Similarly, SWCNT or MWCNT failed to
activate NO, TNF- or IL-8 in A549 cells or NR8383 rat alveolar
macrophages (Pulskump et al., 2007). Likewise, SWCNT failed to
generate intracellular ROS in NR8383 cells after removal of contaminating iron (Pulskump et al., 2007).

Numerous epidemiologic studies have provided evidence that air


pollution contributes to pulmonary and cardio-vascular systemic
morbidity and mortality in susceptible populations (Pope, 2000;
Walker & Mouton, 2008; Sint et al., 2008). Similarly, adverse health
effects may arise from occupational and environmental exposure to
engineered nanomaterials. Health effects and occupational risk of
exposures associated with manufacturing and application of nanoparticles are critical points to consider for safe and sustainable
development of nanotechnology and other emerging technologies
(Maynard et al., 2006). The toxic effects of nanoscale materials have
not been fully characterized and the limited in vivo studies conducted
to date indicate the urgent necessity for further toxicological
assessments of nanomaterials, especially in light of the possible use
of nanomaterials (including CNT) for monitoring and treatment of
human disease.

2.4. Interference with bacterial uptake and killing

3.1. SWCNT in vivo toxicity

Shvedova et al. (2008a) have shown that pre-exposure to SWCNT


decreases the ability of the mouse lung to recover from a bacterial
infection. In vitro evidence indicates that SWCNT inhibit the ability of
RAW 264.7 cell to produce cyclooxygenase-2 in response to the bacterial
lipopolysaccharide (Dutta et al., 2007). Shvedova et al. (2008a) reported
that in vitro exposure of primary mouse alveolar macrophages to
SWCNT decreases phagocytosis and NO production in response to
bacteria. Likewise, SWCNT or MWCNT have been shown to decrease
phagocytosis of latex beads by guinea pig alveolar macrophages in vitro
(Jia et al., 2005). For comparison, pre-incubation with SWCNT was also
found to suppress subsequent uptake of apoptotic target cells by
primary human macrophages (Witasp et al., manuscript in preparation).

Several recent studies showed that the toxicity of inhaled


nanoparticles depends on their physical properties, e.g., particle size
and size distribution, high surface area, and reactivity that also could
affect and facilitate distribution and deposition particles within the
lung compartments (Oberdrster et al., 1994; Oberdrster, 1996;
Donaldson et al., 2006). However, studies of in vivo effects of SWCNT
are still scarce. At the time of writing, a few published reports have
demonstrated the pulmonary toxicity of SWCNT (Warheit et al., 2003;
Lam et al., 2004; Shvedova et al., 2005, 2008b; Li et al., 2007). The IT
instillation or aspiration of SWCNT revealed formation of granulomatous lesions encased by epithelioid macrophages produced by large
agglomerates (Warheit et al., 2003; Lam et al., 2004; Shvedova et al.,
2005). An unusual inammatory response to SWCNT delivered to the
lung via pharyngeal aspiration was characterized by a brisk acute
phase inammatory response followed by an early onset of lung
brosis (Shvedova et al., 2005, 2008b). A well dispersed preparation of
SWCNT (DSWCNT) with a mean diameter of 0.69 m given by
pharyngeal aspiration to C57BL/6 mice (10 g/mouse) revealed a
better interstitial distribution of DSWCNT deposits (b1 m) by 1 day
post-exposure (Mercer et al., 2008). Macrophage phagocytosis of
dispersed SWCNT was rarely observed at any time point (up to
1 month). No granulomatous lesions or epithelioid macrophages were
detected after administration of DSWCNT. However, lung sections and
patterns of cells recovered in lavage uid demonstrated an early,
transient (rapidly resolved) neutrophilic inammatory response that
was very similar to that observed with large SWCNT agglomerates
(Shvedova et al., 2005). Apparently, dispersed SWCNT are rapidly
incorporated into the alveolar interstitium producing an accelerated
increase in collagen deposition. Mercer et al. (2008) emphasized that
there are at least three principal pathways for dissemination of
SWCNT within the lung. Metal-triggered radial self-assembly of
collagen peptide bers was recently demonstrated (Przybyla &
Chmielewski, 2008). In particular, the assembly of the bers was
made possible and triggered by the addition of iron [Fe(II)].
Approximately 80% of the less dispersed material deposited in the
lung is represented by large agglomerates forming granulomatous
lesions in the alveolar region immediately proximal to the small
airways. The remaining less dispersed nanotubes appear to be
aspirated as smaller structures and/or are dispersed by the lung
milieu into a ner form. This fraction of SWCNT is widely distributed
throughout the airspaces of the lung parenchyma and is rapidly
incorporated into the alveolar interstitial wall inducing increased

2.5. Stimulation of lung broblasts


In a mouse model, Shvedova et al. (2005) reported that aspiration
of SWCNT caused diffuse interstitial brosis of rapid onset which
progressed in the absence of persistent inammation. In vitro studies
indicate that lung broblasts may be uniquely sensitive to SWCNT.
Kagan et al. (2007) found that in vitro exposure of lung broblasts
induced expression of mRNA for GM-GSF, IL-1, IL-6, and IL-10 to a far
greater extent than in alveolar type II epithelial cells or alveolar
macrophages. Furthermore, exposure of lung broblasts to SWCNT in
vitro resulted in a 70% increase in cell proliferation and a several fold
increase in collagen production (Wang et al., 2008). Such activation of
lung broblasts may explain the in vivo brogenic reaction to SWCNT
exposure (discussed below).
2.6. Activation of mesothelial cells
Recently, Poland et al. (2008) reported that abdominal injection of
long but not short MWCNT caused an inammatory reaction of the
abdominal lining, which was similar to that seen after exposure of
mice to asbestos. Furthermore, recent in vitro studies support the fact
that mesothelial cells are sensitive to CNT exposure. Exposure of
mesothelial cells in cell culture to SWCNT high in Ni contamination
caused DNA damage, phosphorylation of H2AX and activation of AP-1,
NF-kB, and Akt (Pacurari et al., 2008b). Similarly, in vitro exposure of
mesothelial cells to MWCNT low in metal contamination resulted in
DNA damage, phosphorylation of H2AX and activation of PARP, AP-1,
and NF-kB (Pacurari et al., 2008a). The signicance of these in vitro
responses to human responses to CNT is unknown, since no evidence

196

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

collagen production which progresses in the absence of persistent


inammation. As improved dispersal of SWCNT caused aspiration of
smaller structures that readily enter the alveolar walls, the pulmonary
response to DSWCNT enhanced the interstitial brosis in the absence
of granulomatous formation (Mercer et al., 2008).
3.2. Inhalation route of exposure versus aspiration
Some argue that IT or pharyngeal aspirationa single exposure to a
bolus of SWCNTis an articial exposure and that the bolus exposures
substantially contribute to the pulmonary response (Warheit et al.,
2003). Moreover, aspiration/intratracheal studies reported thus far
have involved relatively high dose exposures, which may not be
relevant to chronic lower dose seen in occupational settings.
Inhalation of SWCNT more closely mimics occupational and environmental venues than the above-mentioned administrations providing
more dispersed SWCNT structures while bolus effects are avoided.
It has been recently reported that application of specially designed
aerosolization technology resulted in the ability to obtain stable
SWCNT aerosols with a concentration of 5 mg/m3 with a count mode
aerodynamic diameter of 240 nm (Baron et al., 2008; Shvedova et al.,
2008b). The inhalation of non-puried SWCNT (iron content of 17.7%
by wt) at 5 mg/m3, 5 h/day for 4 days was compared with pharyngeal
aspiration of varying doses (520 g/mouse) of the same SWCNT in
this study. The chain of pathological events in both exposure routes
was realized through qualitatively similar synergized interactions of
early inammatory response and oxidative stress culminating in the
development of multifocal granulomatous pneumonia and interstitial
brosis (Fig. 2). Quantitatively, SWCNT inhalation was more effective
than aspiration in causing inammatory response, oxidative stress,
collagen deposition and brosis as well as mutations of K-ras gene
locus in the lung of C57BL/6 mice (Shvedova et al., 2008b). Observed
toxic outcomes of exposure of C57BL/6 mice to aerosolized respirable
SWCNT (5 mg/m3, 5 h, 4 days) resulted in a calculated lung burden of
5 g/mouse. This lung burden would be achieved by workers exposed
for less than 1 year at the peak airborne concentrations measured in
an occupational setting based on data of Maynard et al. (2004). Under
these conditions the amount of material produced was very small
(several grams), signicant care was taken to reduce product loss
during handling, and the observed airborne particle concentrations
were low (with peak exposure of 53 g/m3). Agitation of the material
by vortexing generated higher levels of respirable SWCNT material.
Thus, under large-scale manufacturing conditions, the potential for
occurrence of occupational exposure may exist. The exposure
concentration used in SWCNT inhalation studies, is the same as the
current permissible exposure level (PEL) set by OSHA for respirable
synthetic graphite dust, i.e., the PEL currently applied for SWCNT.
Based on the outcomes of SWCNT inhalation study, it could be inferred
that if workers were subjected to long-term exposures to respirable
SWCNT at the current PEL for synthetic graphite, they would likely
have increased risk for pulmonary changes. These results may also be
compared with regulatory permissible levels established for ambient
micro-particles, such as EPA standard for PM2.5 which is 65 g/m3 24 h
and 15 g/m3 annual average (EPA, 2006).

mice (Shvedova et al., 2008a). Decreased bacterial clearance in


SWCNT-pre-exposed mice was associated with decreased phagocytosis of bacteria by macrophages and a decrease in NO production by
these phagocytes. Pre-incubation of nave alveolar macrophages with
SWCNT in vitro also resulted in decreased nitric oxide generation and
suppressed phagocytizing activity toward LM. Failure of SWCNTexposed mice to clear LM led to a continued elevation in nearly all
major chemokines and acute phase cytokines into the later course of
infection. In SWCNT/LM-exposed mice, bronchoalveolar lavage neutrophils, alveolar macrophages, and lymphocytes, as well as lactate
dehydrogenase level, were increased compared with mice exposed to
SWCNT or LM alone. Exposure to carbon nanotubes and infection
induces unusual responses which both mutually enhance inammation and depress bacterial clearance. Thus, exposure to carbon
nanotubes through inhalation may exacerbate lung infection in
susceptible populations.
3.4. Inammatory factors and oxidants
The lung is the prime target for oxidative injury. Numerous studies
have documented that the inammatory response to different
chemicals and pathogens is intrinsically intertwined with simultaneously evolving oxidative stress (Li et al., 2008). SWCNT induced
oxidative stress is one of the important mechanisms contributing to
pulmonary toxicity. SWCNT cause robust inammatory responses in
rodents with very early termination of the acute phase and onset of
chronic brosis. Timely elimination of PMNs through apoptosis and
their subsequent clearance by macrophages is a necessary stage in the
resolution of pulmonary inammation, whereby NADPH oxidase
contributes to control of apoptotic cell death and clearance of PMNs
(Fadeel & Kagan, 2003). However, this role of NADPH oxidaseas a
regulator of the transition from the acute to the brotic stage of
inammationis likely associated with enhanced oxidative stress. In
NADPH oxidase-decient mice which lack the gp91phox sub-unit of the
enzymatic complex, the responses to SWCNT exposure were characterized with a marked accumulation of PMNs and elevated levels of
apoptotic cells in the lungs, production of pro-inammatory cytokines
as well as decreased production of the anti-inammatory and probrotic cytokine, TGF-, and signicantly lower levels of collagen
deposition (Shvedova et al., 2008c). This emphasizes a signicant role
for NADPH oxidase-derived reactive oxygen species in determining
the susceptibility to SWCNT-induced pulmonary damage.
The importance of oxidative stress in pulmonary inammation is
strongly supported by a signicant number of studies in which several
antioxidantsmainly precursors of glutathione, such as N-acetyl-Lcysteine (Sadowska et al., 2006), or superoxide dismutase (SOD) and

3.3. Interference with bacterial uptake


Realistic exposures to SWCNT may occur in conjunction with other
pathogenic impacts (microbial infections) and trigger enhanced
responses. Evaluated interactions between pharyngeal aspiration of
SWCNT and bacterial pulmonary infection of C57BL/6 mice with Listeria monocytogenes (LM) have shown that sequential exposure to
SWCNT/LM amplied lung inammation and collagen formation.
Despite this robust inammatory response, SWCNT pre-exposure
signicantly decreased the pulmonary clearance of LM-exposed mice
measured 3 to 7 days after microbial infection versus PBS/LM-treated

20 m

Fig. 2. Representative image of lung section from the SWCNT inhalation study (5 mg/m3,
5 h/day, 4 days) depicting granuloma formation on day 28 post treatment. Fibrosis is
indicated by blue staining in this Masson's Trichrome stained section of the lung.

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

SOD mimeticshave been successfully employed as protective antiinammatory agents (Salvemini et al., 2002; Kinnula & Crapo, 2004).
Surprisingly, only few investigations addressed the role of vitamin E in
pulmonary inammatory response (Singh et al., 2005). It was reported
that vitamin E supplementation decreased the number of neutrophils
during inammatory response (Rocksn et al., 2003) but increased
recruitment of macrophages (Belo et al., 2005). In C57BL/6 mice
maintained on a vitamin E-decient diet and treated with SWCNT,
more severe decrease of pulmonary antioxidants was detected as
compared to vitamin E-sufcient SWCNT-treated controls (Shvedova
et al., 2007a). Exposure to SWCNTs markedly shifted the ratio of
cleaved to full length extracellular SOD. The ratio of cleaved (lacking
matrix binding domain) to full-length extracellular SOD was 34
times greater in SWCNT exposed mice compared to control mice.
Lowered levels of antioxidants in vitamin E-decient mice were
associated with a higher sensitivity to SWCNT-induced acute
inammation (total number of inammatory cells, number of PMNs,
released lactate dehydrogenase, total protein content, and levels of
pro-inammatory cytokines, TNF-a and IL-6) as well as pro-brotic
pathways (elevation of TGF- and collagen deposition). Given that
pulmonary levels of vitamin E can be manipulated through diet, its
effects on SWCNT responses may be of practical importance in
optimizing protective anti-inammatory strategies.
4. In vivo evaluation of the
bioactivity of multi-walled carbon nanotubes
The potential for occupational exposures to different types of CNT
SWCNT and MWCNT exits throughout their life cycle, and in particular
inhalation exposure to airborne CNT is of major occupational and
environmental concern (Maynard & Kuempel, 2005), but very little is
currently known about these exposures. A recent study reported
MWCNT-containing airborne dust levels from undetectable to
~400 g/m3 in a research laboratory (Han et al., 2008). The levels varied
with different operations involving MWCNT, i.e., spraying, blending, or
weighing produced the highest exposure concentrations, but the total
mass concentration reported in this study was not comprised
exclusively of MWCNT. Implementation of controls signicantly reduced
airborne concentrations, often to non-detectable levels.
4.1. MWCNT and ROS generation
A hierarchical oxidative stress model that predicts engineered
nanoparticle toxicity has been proposed (Nel et al., 2006). Data on the
applicability of this hierarchical oxidative stress model to MWCNT is
currently a matter of debate. Cell-free experiments using MWCNT in
aqueous suspension determined that MWCNT do not directly generate
either oxygen or carbon-centered free radicals (Fenoglio et al., 2006).
In contrast, in vitro studies with NR8383 and A549 cells demonstrated
dose- and time-dependent increases in intracellular ROS production
after exposure to SWCNT or MWCNT, suggesting that CNT exposure
induces cellular oxidative stress (Pulskump et al., 2007). However,
when the carbon nanotubes used in these experiments were acidtreated to remove metal contaminates, the cellular ROS generation did
not occur, suggesting the metal catalyst contaminates were responsible for the ROS generation.
4.2. MWCNT in vivo toxicity studies
In vivo studies of MWCNT are very limited, and the results of these
studies are not consistent. The pulmonary toxicity of MWCNT in rats
exposed by IT instillation to whole (5.9 m long) or ground (0.7 m
long) MWCNT reported both MWCNT samples caused acute pulmonary inammation at 3 and 15 days post-exposure, and induced
pulmonary brosis at 60 days post-exposure (Muller et al., 2005).
Inhalation exposure of mice to MWCNT has been reported to induce

197

no signicant lung toxicity, but did cause immunosuppression


(Mitchell et al., 2007). The differential results of these two studies
have been attributed to differences in MWCNT surface area, rodent
species used, and differences in observation period (McDonald &
Mitchell, 2008). Alternatively, it has been suggested that the
nanoparticles used in the inhalation study (Mitchell et al., 2007)
were carbon stacked cup nanobers, not MWCNT (Lison & Muller,
2008). Ongoing studies in our laboratory contribute further to the
controversy surrounding the in vivo toxicity of MWCNT. Specically,
our studies have determined that aspiration exposure of mice to
MWCNT resulted in dose- and time-dependent pulmonary inammation and damage, as well as effects in the central nervous system
(Sriram et al., 2007). In addition, preliminary results of our MWCNT
inhalation study indicate that inhaled MWCNT induce a dosedependent pulmonary inammation and damage in mice consistent
with our aspiration study results (Porter, unpublished observations).
Interestingly, very recent studies reported by Fenoglio et al. (2008)
and Muller et al. (2008) suggest that structural defects may play a
major role in the acute lung toxicity of multi-walled carbon
nanotubes. To examine how structural properties may modulate the
toxicity of CNT, these investigators used preparations of MWCNT that
had been modied either by grinding (thereby introducing structural
defects) and subsequently heated either in a vacuum at 600 C
(causing reduction of oxygenated carbon functionalities and reduction
of metallic oxides) or in an inert atmosphere at 2400 C (causing
elimination of metals and annealing of defects), or by heating at 2400
in an inert atmosphere and subsequently grinding the thermallytreated CNT (introducing defects in a metal-deprived carbon framework). The studies suggested that the acute pulmonary toxicity
following IT administration of MWCNT (2 mg/animal) to Wistar rats,
as well as the genotoxicity of MWCNT, assessed in vitro using
immortalized rat lung epithelial cells, was mitigated upon heating
but restored upon grinding, indicating that the intrinsic toxicity of
MWCNT is mediated, at least in part, by the presence of defective sites
in their carbon framework.
In sum, it is likely that many physico-chemical characteristics of
MWCNT, e.g., length, width, structural defects, metal contamination,
surface chemistry, and zeta potential, will affect the lung response to
MWCNT. Until the physico-chemical parameters of MWCNT which
contribute to their toxicity are uniformly characterized, it is likely that
different types of MWCNT materials will result in differing biological
responses. Thus, until MWCNT used for in vivo studies are extensively
characterized and these physico-chemical differences compared to
lung responses, it will be difcult to reach a consensus on the
pulmonary toxicity of MWCNT.
4.3. MWCNT translocation
Translocation of nanoparticles from the lung to the blood and
extra-pulmonary organs has been documented for various nanoparticles. For example, 60 min after IT instillation of hamsters with
nanocolloid albumin labeled with technetium-99m, detectable levels
of the labeled nanoparticles were found in the blood, liver, heart
spleen kidneys and brain (Nemmar et al., 2001). Rats exposed by
inhalation exposure to 13C-labeled ultrane carbon particles demonstrated that liver accumulated a signicant amount of the ultrane
carbon particles by 24 post-exposure, with little detectable deposition
in other extra-pulmonary organs (Oberdrster et al., 2002). An inverse
relationship between particle size and systemic distribution after
inhalation exposure to 15 and 80 nm iridium particles has also been
reported (Kreyling et al., 2002). Humans exposed by inhalation to
technetium-99m-labeled ultrane carbon particles determined that
the labeled carbon particles were detected in the blood within 1 min
after exposure, reached constant levels between 10 and 20 min of
exposure, and remained at this level for 60 min (Nemmar et al., 2002).
Substantial amounts of labeled nanoparticles were found throughout

198

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

various organs as well (Nemmar et al., 2002). Importantly, the latter


study indicated that nanoparticle translocation can occur in humans,
and should be considered when assessing nanoparticle toxicity.
To date, only one study has examined the translocation of MWCNT.
In this study, mice were exposed to 14C-taurine-labeled MWCNT by
three exposure routes; intravenous injection, IT instillation, and
gavage (Deng et al., 2007). Within 10 min after intravenous injection,
14
C-taurine-labeled MWCNT accumulated in the liver, heart and lung,
but not other organs tested, including brain, stomach, muscle, bone,
and intestine. The liver was the predominant site of deposition, with
80% of the exposed dose, which remained relatively unchanged for
28 days post-exposure. By 90 days post-exposure, the amount of 14Ctaurine-labeled MWCNT in the liver decreased to 20% of the exposed
dose. After gavage exposure, 14C-taurine-labeled MWCNT was only
detectable in the stomach, small intestine, large intestine and feces.
No 14C-taurine-labeled MWCNT was detected in the blood. After IT
exposure, 14C-taurine-labeled MWCNT levels in the lung decreased
from 78% to 20% of the total exposed dose from 1 day to 28 days postexposure, indicating the mouse lung can clear MWCNT. However, no
14
C-taurine-labeled MWCNT was found in blood or other major organs
after IT exposure. The ndings of this initial study on MWCNT
translocation, which used 14C-taurine-labeled MWCNT, could thus
offer our rst insight into the potential translocation of MWCNT.
However, these investigations may be confounded by the label used
for monitoring the MWCNT. The physiology and biochemistry of
taurine are well documented and have been reviewed elsewhere
(Wright et al., 1986). Specically relevant to the Deng et al. (2007)
study is the fact that high afnity taurine uptake systems exist in
various cell and tissue types in rodents and humans. It is difcult to
rule out the possibility that these cellular taurine uptake systems may
have contributed to facilitating the uptake of 14C-taurine-labeled
MWCNT, and this may in turn have impacted the distribution and
clearance of the MWCNT.
4.4. MWCNT and induction of mesothelioma
Since MWCNT exhibit a high dimensional aspect ratio, i.e.
exhibiting a long and thin morphology, questions concerning
asbestos-like pulmonary responses have been raised. Two recent
studies of the responses of mice to intra-abdominal injection of
MWCNT have received a great deal of attention in the lay press. The
rst study reported that mesothelioma was observed 6 months after
intraperitoneal injection in both the MWCNT-exposed and the
asbestos-exposed mice, but not in the fullerene-exposed mice (Takagi
et al., 2008). In addition, the death rate for the MWCNT-exposed group
was signicantly greater than the asbestos-exposed group. Of note,
MWCNT were administered to mice heterozygous for the tumor
suppressor, p53 that have been reported to be especially sensitive to
asbestos-induced mesothelioma (Vaslet et al., 2002). Although the
ndings of Takagi and colleagues are striking, the study has been
criticized for the use of an extraordinarily high (3 mg/mouse)
exposure dose (Donaldson et al., 2008; Ichihara et al., 2008). This
high exposure dose may have resulted in extensive abdominal brosis
causing death by constriction ileus. Recently, Poland et al. (2008)
reported an acute response to intra-abdominal instillation of a more
reasonable dose (50 g/mouse) of MWCNT. They report that long
MWCNT but not short MWCNT caused inammation of the abdominal
wall, with formation of so-called foreign body giant cells, and an
increase in polymorphonuclear leukocytes harvested by peritoneal
lavage at seven days post-exposure. These responses to long MWCNT
were similar in magnitude to asbestos-induced responses. Issues
which remain are whether this acute inammatory response would
persist and progress to mesothelioma (there is a lag-time of up to
30 years for mesothelioma formation in humans exposed to
pathogenic asbestos bers) and whether inhaled MWCNT would be
able to migrate to the intrapleural space and affect the mesothelium. It

is clear that more toxicological research is needed to determine


exactly what type of carbon nanotubes pose the greatest risk, and
whether mesothelioma, or other lung diseases, are possible outcomes
after human exposure to these novel materials.
5. Genotoxicity and mutagenicity of carbon nanotubes
Deep penetration of nanotechnologies and nanomaterials into
essentially all spheres of society and increasing numbers of carbon
nanotube-containing consumer products raise concerns about their
possible genotoxic effects (Kagan et al., 2005; Donaldson, 2006; Roco,
2006; Singh & Nalwa, 2007). Moreover, the studies cited above on the
potential asbestos-like pathogenicity of CNT also add to the concern
that these materials may present a carcinogenic risk. Inammation
and pulmonary brosis have been associated with an increased risk
for lung cancer (Hubbard et al., 2000; Knaapen et al., 2004) thus
justifying assessments of genotoxic events possibly accompanying
CNT exposure. However, in vivo and in vitro genotoxicity studies with
CNT are very limited, and the results obtained to date are not
consistent. Kisin et al. (2007) examined the genotoxic effects of
puried SWCNT (0.23 wt.% Fe) using three different test systems: the
comet assay and micronucleus (MN) test in a lung broblast (V79) cell
line, and the Salmonella gene mutation assay in YG1024/YG1029
strains. The genotoxicity analysis showed a SWCNT-concentrationdependent increase in the frequency of DNA damage, in terms of
strand breaks, but did not produce signicant micronucleus induction
in Chinese hamster lung broblast cells. The results clearly indicated
that SWCNT-exposed V79 cells displayed a signicant time- and
concentration-dependent loss of viability. This suggests that SWCNT
induced loss of cells might interfere with accurate evaluation of
genotoxicity responses to SWCNT detectable in viable cells. Further,
the negative response in the MN assay may be due, at least in part, to a
low degree of SWCNT uptake by V79 cells. Indeed, Kagan et al. (2006)
reported that SWCNT are not particularly active targets of macrophage
phagocytosis. Moreover, no increased mutation frequencies in either
YG1024 or YG1029 strains of Salmonella typhimurium were detected at
all concentrations of SWCNT employed in the study. A pilot study
performed by Szendi and Varga (2008) showed that single oral
exposure to SWCNT (90% purity) or MWCNT (9598% purity) did not
increase urinary mutagenicity in the rats. Moreover, no signicant
genotoxic effects were found in the human lymphocytes after single
exposure to MWCNT by MN and sister chromatid exchange assays.
SWCNT genotoxicity analyses were not performed due to the high
cytotoxic concentration (1 mg/ml) used in the study. Jacobsen et al.
(2008) assessed genotoxicity and mutagenicity of SWCNT (2% Fe) in
the FE1-MetaMT mouse lung epithelial cell line. SWCNT exposure did
not induce any increases in cII mutant frequency or in the level of
strand breaks, but resulted in oxidation of purines as detected by
signicant generation of formamidopyrimidine [fapy]-DNA glycosylase sensitive sites. The induction of formamidopyrimidine [fapy]DNA glycosylase sites corresponded to 630 lesions per diploid cell
after treatment with SWCNT. Indeed, DNA base oxidation has been
found to be much more sensitive than strand breaks to environmental
factors (Vinzents et al., 2005). On the other hand, Zhu et al. (2007)
demonstrated the DNA damage and increased mutation frequency in
mouse embryonic stem cells in response to MWCNT exposure.
Furthermore, Karlsson et al. (2008) showed a signicant increase in
DNA damage caused by MWCNT in A549 type II epithelial cells, but no
oxidative DNA lesions. Additionally, as mentioned above, intraperitoneally-administered MWCNT induced mesothelioma in p53 heterozygous (+/) mice (Takagi et al., 2008). The p53 (+/) mice, in
general, have been suggested to be a good model to predict
carcinogenicity (Pritchard et al., 2003). Although the genotoxic effect
of MWCNT was unclear, MWCNT exerted carcinogenic potential in
p53 (+/) mice which was presumably dependent on the size/shape
and persistence in the organism.

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

199

Recently, Shvedova et al. (2008b) demonstrated that SWCNT


inhalation resulted in mutations of K-ras gene locus in the lung of
C57BL/6 mice. Among the mutated genes implicated in pulmonary
tumorigenesis, the K-ras oncogene is frequently found in lung tumors
of mice exposed to chemicals (Devereux et al., 1993; Ronai et al., 1993).
The increased rate of mutations was found very early (on days 17)
after inhalation of non-puried SWCNT (17.7 wt.% Fe) and persisted
throughout day 28. The effect was coincident with the time of
maximal inammatory response, suggesting that inammation and
resultant oxidative injury may be a cause of mutagenicity. Given that
the oxidative burst and accompanying oxidative stress are known to
be signicant contributors to genotoxicity, it is tempting to speculate
that there is a causative association between accumulation of K-ras
mutations and oxidative stress triggered by SWCNT inhalation. The
types of mutations consisted of G to A transition that changed the wild
type codon 12 of the K-ras gene from GGT (glycine) to either AGT
(serine) or GAT (aspartate). This type of mutation has been reported
previously in lung tumors from susceptible mice (Devereux et al.,
1993; Ronai et al., 1993). For instance, lung tumors in mice exposed to
the lung carcinogen, NNK [4-(methylnitrosamino)-1-(3-pyridyl)-1butanone], showed a high proportion of this mutation that has been
suggested to occur via the mutagenic lesions, including O6-methylguanine, induced by NNK metabolites (Devereux et al., 1993).
Interestingly, one of the mutations found in SWCNT-treated mice (at
day 28 post SWCNT inhalation) consisted of a double mutation
occurring at codons 12 and 8 [GGT to GAT and GTG to ATG (valine to
methionine), respectively]. The role of this double mutation is
unknown and may be specic for SWCNT exposure. It was proposed
that brosis might be involved in the carcinogenesis by the occurrence
of atypical or dysplastic epithelial changes which progressed to
invasive malignancy (Fraire & Greenberg, 1973). However, the
involvement of K-ras mutations in brosis-associated lung cancer is
not understood and merits further investigation.
Taken together, studies of the carcinogenic potential of carbon
nanotubes are still in the early phases, but some of the results reported
to date suggest that one should proceed with caution. However, this
does not necessarily entail that carbon nanotubes should be
abandoned altogether in technological or biomedical applications, as
risk is always the combination of both hazard and potential for
exposure (Maynard et al., 2006; Stern &McNeil, 2008). More studies
on environmental and occupational exposure to engineered nanomaterials are thus needed.

date, only a few studies have been published concerning the


biodistribution of CNT in animals. Wang et al. (2004) used 125Iodinelabeled hydroxylated SWCNT (125I-SWCNT-OH), functionalized by
oxidation of the nanotubes, and monitored their distribution in mice
following various routes of administration. These authors reported
that the biodistribution of the functionalized, water-soluble CNT was
not signicantly inuenced by the administration route and that the
125
I-SWNT-OH distributed rapidly throughout the body, and were
then excreted with the urine and feces. A second study focused on the
intravenous route of administration of functionalized SWCNT and
MWCNT using a different surface chemistry (Singh et al., 2006). The
CNT were functionalized with the chelating molecule DTPA and
radiolabeled with 111indium (111In-DTPA-CNT). These studies indicated that functionalized 111in-DTPA-CNT is not retained in any of the
reticuloendothelial system organs (liver or spleen) and were rapidly
cleared from systemic blood circulation through the renal excretion
route. Moreover, electron microscopy analysis of urine samples
revealed that both types of nanotubes (SWCNT and MWCNT) were
excreted as intact nanotubes. A more recent study utilized the intrinsic
Raman spectroscopic signatures of SWCNT, to monitor the blood
circulation of intravenously injected SWCNT and to detect SWCNT in
various organs and tissues of mice ex vivo over a period of 3 months
(Liu et al., 2008). These authors found that functionalization of SWCNT
by branched polyethylene-glycol chains enabled prolonged SWCNT
blood circulation (up to 1 day), relatively low uptake in the
reticuloendothelial system, and near-complete clearance from major
organs in ~2 months. Furthermore, no toxicity of SWCNT to mice was
observed in necropsy, histology, and blood chemistry measurements.
Using a similar strategy, Liu et al. (2007) have shown that SWCNT can
be targeted to integrin-positive tumors in mice through the attachment of arginine-glycine-aspartic acid (RDG) peptides to polyethylene-glycol chains coated onto the surface of SWCNT.
Overall, it appears that the surface chemistry of carbon nanotubes is
critical to their in vivo behavior and distribution, a conclusion that may
well apply to other engineered nanomaterials. This is expected because
pristine carbon nanotubes have very hydrophobic surfaces. Indeed, it has
recently been shown that intravenously-injected pristine (non-functionalized) SWCNT is highly enriched in liver, lungs, and spleen in mice and
remains in the body over an extended period of time (Yang et al., 2007).
Controlling the biodistribution of carbon nanotubes through surface
functionalization will thus be instrumental in transforming these
materials into candidates for delivery of pharmaceutical agents.

6. Perspectives on medical applications of carbon nanotubes

6.2. CNT for cancer treatment

The above described potent inammatory responses synergized


with oxidative stress induced by CNT in the lung may generate an
impression that their medical applications should be limited, if
possible at all. Indeed, toxic interactions with different cell populations, brosis-causing capacities, asbestos-like activities, bioaccumulation and potentially low levels of biodegradation and other
threatening consequences dampen the enthusiasm towards using
CNT as diagnostic tools and as vehicles for drug delivery. However, this
is primarily relevant to non-functionalized CNT. Several recent
developments, particularly in synthesis of chemically-modied CNT
resulting in their enhanced solubilization (dispersibility) and
improved biocompatibility makes them more versatile and shows
potential pathways for their utilization in medicine (Lacerda et al.,
2006; Prato et al., 2008). The following sections will provide a brief
overview of some recent approaches taken in the eld of carbon
nanotube development in the pharmaceutical eld.

Due to their strong optical absorbance, carbon nanotubes have


become candidates for biophysical approaches to destroy cancer cells
using, for example, hyperthermia. In principle, functionalization of
CNT with tumor-specic epitopes followed by uptake only by
cancerous cells affords an opportunity to locally heat the carbon
nanotubes using infrared laser radiation, resulting in thermal destruction of cancer cells without harming surrounding cells. Using this
approach, Kam et al. (2005) have shown that SWCNT functionalized
with folic acid were specically internalized by cancer cells (in vitro)
expressing folic acid receptors on their surface. Hyperthermic
treatment using 700- to 1100-nm near-infrared light resulted
in killing of cancer cells without harming receptor-free normal cells.
Hence, the strong optical absorbance of SWCNT in this special spectral
window, an intrinsic property of this material, can be used for selective optical stimulation inside cells. Similar approaches to thermal
ablative cancer therapy (in vitro and in vivo) have been taken using
gold (Au) nanoshells that were tuned to strongly absorb light in the
near infrared (Hirsch et al., 2003).
An alternative strategy for tumor targeting of carbon nanotubes
was demonstrated recently by McDevitt et al. (2007), who reported on
the functionalization of carbon nanotubes with monoclonal

6.1. Biodistribution of functionalized CNT


The fate and distribution of carbon nanotubes upon in vivo
administration are critical to potential applications in medicine. To

200

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

antibodies. Tumor-targeted SWCNT constructs were synthesized from


functionalized, water-soluble carbon nanotubes by covalently attaching the tumor-specic monoclonal anti-CD20 antibody (Rituximab),
radiometal-ion chelates, and uorescent probes. The 111In-labeled and
Rituximab-functionalized SWCNT specically localized in disseminated human B-cell lymphoma xenografts in the bone marrow and
spleen in mice, after intravenous injection. These results suggest that
carbon nanotubes may be useful vehicles for transporting radionuclides to malignancies, although more information is needed on
potential risks of CNT as delivery systems.
Finally, using a combination of antibody-directed targeting and
thermal ablation, Chakravarty et al. (2008) have recently shown
that SWCNT can be used for selective killing of human cancer cells.
These authors used anti-CD22 antibodies to target B lymphoma cells
in vitro followed by exposure of these cells to laser radiation. It will, of
course, be of interest to learn more about the pharmacokinetics, biodistribution, toxicity, and activity of the novel monoclonal antibodyCNT constructs in vivo.
6.3. CNT for infectious diseases
Internalization of carbon nanotubes by cells is also a critical determinant in different biomedical applications such as delivery of
therapeutically active molecules, including proteins, peptides, and
genes. As discussed in previous sections, pristine carbon nanotubes
are not readily recognized by professional phagocytes of the immune
system. However, recent studies have shown that functionalization of
SWCNT and MWCNT using the 1,3-dipolar cycloaddition reaction that
allows insertion of amino functions onto the CNT, thus rendering the
carbon nanotubes water-soluble, allows for the internalization of CNT
by a wide range of cell types, albeit in a non-specic manner
(Kostarelos et al., 2007; Zhang et al., 2007). Enhanced cellular uptake
of SWCNT was also reported when carbon nanotubes were coated
with an amphiphilic helical peptide (Chin et al., 2007). The mechanism of cellular uptake of CNT has been debated (Pantarotto et al.,
2004; Kam et al., 2006), but should be claried to allow for targeted
delivery of carbon nanotubes and their cargo.
Our own recent work has identied PS-functionalization as a
potentially useful approach to target professional phagocytes
(macrophages) with SWCNT (Fig. 1) (Konduru et al., submitted for
publication). PS is an endogenous recognition signal that is also
utilized by apoptotic cells and aging red blood cells (Witasp et al.,
2008) and thus feeds into well-controlled signaling pathways in
phagocytic cells orchestrating their behavior, including secretion of
cytokines. Macrophage uptake of PS-functionalized SWCNT was
shown to be time-dependent and was blocked by pharmacological
inhibitors of endocytosis; moreover, uorescently labeled PS-SWCNT
were found to co-localize with markers of the endo-lysosomal
compartment, further supporting the notion that uptake was specic
and mediated through endocytosis. In addition, our in vitro studies
have shown that PS-coated SWCNT can function as vectors for the
delivery into macrophages of pro-apoptotic factors, such as cytochrome c, resulting in killing of cells through activation of the
cellular apoptosis program (Konduru et al., submitted for publication). Selective targeting of CNT with lethal cargoes to macrophages
harboring intracellular pathogens could represent a signicant
therapeutic advantage when traditional anti-microbial remedies
are ineffective. Of course, animal studies are required to test the
feasibility and effectiveness of this approach.
Another potential therapeutic approach is to use functionalized
CNT for immune activation and/or vaccination. Pantarotto et al. (2003)
reported that the covalent attachment of a neutralizing B cell epitope
from the foot-and-mouth disease virus to CNT elicited strong antipeptide antibody responses in mice with no detectable crossreactivity to the carbon nanotubes. The fact that a biologically relevant
epitope was presented in an appropriate conformation is promising

and suggests that functionalized carbon nanotubes could be utilized


for immunization purposes, if toxicity is controlled.
6.4. CNT as imaging agents and sensors
Carbon nanotubes have a tunable near-infrared emission that
responds to changes in the local dielectric function but remains stable
to permanent photobleaching. Barone et al. (2005) reported the
synthesis of solution-phase, near-infrared sensors, with -D-glucose
sensing as a model system, using SWCNT that modulate their emission
in response to the adsorption of specic biomolecules. By inserting a
capillary tube containing such functionalized nanotubes into human
epidermal tissue (ex vivo), they could show that the carbon nanotube
senses glucose concentration in the range commonly found in the
blood of diabetic patients (18 mM). CNT could thus potentially serve
as biosensors, as they emit uorescence in the near infrared, a region
of the spectrum not occupied by other organic molecules and where
tissue is transparent. Molecular detection using near-infrared light is
relevant in biomedical applications because of greater tissue penetration and reduced auto-uorescent background in human tissue or
whole-blood media.
Another very recent study has shown that carbon nanotubes could
be employed as novel photoacoustic molecular imaging agents.
Photoacoustic imaging has the potential to image animal or human
organs with simultaneous high contrast and high spatial resolution,
and De la Zerda et al. (2008) have provided evidence that SWCNT
conjugated with RDG peptides can be used as a contrast agent for
photoacoustic imaging in live animals (i.e. mice bearing integrinpositive tumors). Importantly, a greater spatial resolution was
achieved by photoacoustic imaging using carbon nanotubes as
compared with uorescence imaging using tumor-targeted quantum
dots. These elegant animal studies suggest new approaches to noninvasive imaging and monitoring of disease processes, taking
advantage of the specic physico-chemical properties of carbon
nanotubes.
7. Regulatory issues related to carbon nanotubes
There are at least ten health and safety laws or regulations in the
United States that could pertain to engineered nanoparticles, including carbon nanotubes. This legislation addresses the environment in
general, including air, water, soil, hazardous waste, as well as uses in
commerce, the workplace, in food, consumer products, pharmaceuticals, and in diagnostic and therapeutic medical devices (Davies,
2006). The dilemma with existing legislation and regulations is
illustrated by the TSCA. The question that arises is whether engineered
nanoparticles, such as carbon nanotubes, should be considered as
new materials under the Act (Davies, 2008). Indeed, a similar
concern is currently being raised with respect to the implementation
of the new REACH (Registration, Evaluation, Authorisation, and
Restriction of Chemicals) legislation in Europe. Some carbon nanotubes may be considered new depending on the EPA interpretation of
the TSCA inventory policy regarding these types of nanoscale
materials. Alternatively, the EPA could utilize authority under TSCA
to consider carbon nanotubes a signicant new use. In either case,
manufacturers would be required to provide data on the toxicity of the
nanomaterial.
The regulatory approach in the United States to hazardous
substances generally has been to consider toxic effects that result
due to chemical composition. Engineered nanoparticles, however, do
not t this general paradigm exactly because such particles, while
often having the same chemical composition as their bulk counterparts, behave differently and have different properties and potential
for toxicity. For example, the most critical regulatory issue that arises
when workers are exposed to carbon nanotubes is that the exposure
limit would be the same as for graphite. The OSHA PEL for graphite is

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

Fig. 3. Carbon nanotubes: engineered nanomaterials with Janus-like properties. Singlewalled or multi-walled carbon nanotubes (CNT) have been reported to produce toxic
effects in cell culture as well as in vivo. This picture summarizes some of the known
toxicities of carbon nanotubes. For more detail, consult the text. It should be noted,
however, that the hazardous effects of CNT may, in some cases, be ascribed to residual
metal or other contaminants arising during the production process, or could be caused
by agglomeration related to the method of dispersion of nanotubes. Moreover, certain
pathogenic effects have been ascribed to specic forms of CNT, such as the putative
association between mesothelioma formation and MWCNT with high aspect ratio.
Overall, various physico-chemical properties of CNT including length, width, presence
of structural defects, metal contaminants, surface chemistry, zeta potential, and so
forth, are likely to determine the in vivo responses following occupational or
environmental exposure to these novel materials. On the other hand, CNT are readily
functionalized and this may result in enhanced dispersibility in biological media (i.e.,
CNT can become water-soluble, with improved biodistribution) and improved
biocompatibility, making them versatile and attractive for biomedical applications.
Some selected examples of pharmaceutical and biomedical application of functionalized carbon nanotubes (f-CNT) are depicted in the schematic diagram, and are
discussed in more detail in the text. The Photoshop representation of the two-headed
herm of Janus was produced by Kimberly S. Clough-Thomas (khc7@cdc.gov).

5 mg/m3. It is considered a nuisance dust. However, Shvedova et al.


(2005) estimated that workers exposed to SWCNT at the OSHA PEL for
graphite, would attain an equivalent lung burden to that causing an
adverse effect (brosis) in mice if the workers were exposed to the
amounts used in the study for only 20 days. Moreover, the brotic
response in mouse lung after exposure to carbon nanotubes by
pharyngeal aspiration or intratracheal instillation was much greater
than that of the same mass of carbon black or quartz (Shvedova et al.,
2005; Lam et al., 2006; Donaldson et al., 2006). These nding were
conrmed in a recent inhalation study of SWCNT (Shvedova et al.,
2008b).
Many studies of ne and ultrane particles have shown that on a
per mass basis, nano-sized particles are more hazardous than their
bulk counterparts (Oberdrster et al., 1990, 1994, 2005; Zhang et al.,
2000, 2003; Chen et al., 2006). In addition to particle size and surface
area, various physico-chemical factors including surface reactivity,
solubility, and shape can inuence the toxicity of engineered
nanoparticles (Maynard & Kuempel, 2005; Oberdrster et al., 2005;
Donaldson et al., 2005; Kuempel et al., 2007). However, for a given
material of the same mass and chemical composition, the decrease in
particle size increases the available particle surface to elicit biological
responses. Nanoparticles can also more readily enter cells and cell
organelles, causing adverse effects such as oxidative stress (Oberdrster et al., 1990, 1994; Li et al., 2003), and novel routes of translocation

201

of nanoparticles from the portal of entry to other organs also must be


considered (Oberdrster et al., 2004). Hence, given the potentially
greater toxicity of nanoparticles relative to bulk materials, it may not
be appropriate that exposure to engineered nanoparticles are
controlled in the workplace to the same level as their bulk counterparts. Using quantitative assessment methods, the National Institute
for Occupational Safety and Health found that the risk of lung cancer
from nano-sized titanium dioxide was greater than for the ne-sized
material and recommended an approximately ten-fold lower exposure limit for the nano-sized material (NIOSH, 2005).
Carbon nanotubes including SWCNT and MWCNT are of special
interest and concern regarding their hazard potential because they are
brous, high aspect ratio nanoparticles (HARN). Their nanometer
diameters and micrometer lengths evoke the previous experience
with asbestos. Preliminary investigations support the notion that
HARN may act like asbestos to some extent. The key question that may
drive regulation is whether HARN, such as SWCNT and MWCNT,
actually have asbestos-like properties. It is not known with certainty
whether HARN will behave like asbestos but the current paradigm of
ber pathogenicity does not discriminate between compositions of
long biopersistent bers (Oberdrster, 2007). For example, the two
long biopersistent bers erionite and silicon carbide whiskers are
particularly potent in causing mesothelioma for reasons that are not
easily explained since they were not especially long or more
biopersistent than amphibole asbestos (Merchant, 1990; Seluk et
al., 1992; Johnson et al., 1992; Akiyama et al., 2007). This suggests that
some ber types might possess surface or chemical reactivity over and
above what would be expected for long biopersistent bers. It is not
known if carbon nanotubes could have the potential; however, there
are substantial possibilities to derivatize and change carbon nanotube
surfaces and thus to alter properties that could induce toxicity (Sayes
et al., 2006; Oberdrster et al., 2007). Recent studies by Poland et al.
(2008) and Takagi et al. (2008) have demonstrated that carbon
nanotubes can trigger some similar biological responses as asbestos. It
is likely that in the absence of countervailing evidence that there will
be pressures on regulators to control occupational exposures to
carbon nanotubes (Gitlin, 2008). Whether this will impact other areas
of regulation is not known. There have been reports that generally,
nanotechnology businesses, insurers, and nongovernmental organizations may support sensible regulation in the nanotechnology area so
as to provide ground rules for otherwise unregulated markets
(Davies, 2006; Lloyd's, 2007). Clearly, there are many health and safety
regulatory questions associated with engineered nanoparticles such
as SWCNT and MWCNT which will need answers in the coming years,
to enable a safe and sustainable development of the nanotechnologies.
8. Concluding remarks
In Roman mythology, Janus was the god of gates and doors, of
beginnings, and of endings. Janus was usually depicted with two
heads looking in opposite directions, and was frequently used to
symbolize change and transitions, such as the progression from past to
future, of one condition to another, or of one vision to another. He was
also known as the gure representing time because he could see into
the past with one face and into the future with the other. How does
the mythological gure of Janus relate to the emerging nanotechnologies, and to the exploitation of engineered nanomaterials in
medicine (Fig. 3)? First, as discussed in the present review, singleand multi-walled carbon nanotubes have been ascribed various toxic
effects in vitro and in vivo, and some studies have even indicated
similarities between certain carbon nanotubes (multi-walled, with
high aspect ratio) and pathogenic asbestos bers. On the other hand,
recent studies have also demonstrated that carbon nanotubes could
potentially be harnessed for biomedical purposes, as vectors for drug
delivery, and as imaging agents. The important task that lies ahead
will thus be to determine which physico-chemical characteristics of

202

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

carbon nanotubes are driving the toxic responses, and to utilize this
knowledge in the design of carbon nanotubes that are biocompatible
and safe. Second, we should look to the past and learn from previous
investigations of particles and bers (such as asbestos) that may
inform us on the toxic potential of emerging nanomaterials (Oberdrster et al., 2005). At the same time, we may look to the future and
recognize that nanomaterials, including carbon nanotubes, are
versatile materials that can be readily functionalized, and which
may one day be introduced into the clinic.
Furthermore, it may be prudent to recognize that some of the
unwanted (toxic) responses of carbon nanotubes may, in fact, lead to
desirable outcomes depending on the specic context. For instance,
the controlled induction of carbon nanotube-dependent cellular
toxicity could serve as a novel and effective approach to combat
cancer cells, as discussed herein (for comparison, arsenic is widely
recognized as a poison, yet has been shown in clinical studies to have
efcacy in the treatment of acute promyelocytic leukemia). Overall, a
detailed understanding of the pharmacological and toxicological
properties of carbon nanotubes and a critical and balanced evaluation
of risk/benet ratios is required as the medical applications of these
novel materials are being considered.
Acknowledgments
This study was supported by grants from NIOSH OH008282, NIH
HL70755, NORA 927000Y, NORA 927Z1LU and the 7th Framework
Program of the European Commission (EC-FP-7-NANOMMUNE-214281).

References
Akiyama, I., Ogami, A., Oyabu, T., Yamato, H., Morimoto, Y., & Tanaka, I. (2007).
Pulmonary effects and biopersistence of deposited silicon carbide whisker after 1year inhalation in rats. Inhal Toxicol 19(2), 141147.
Awasthi, K., Srivastava, A., & Srivastava, O. N. (2005). Synthesis of carbon nanotubes.
J Nanosci Nanotechnol 5(10), 16161636.
Barone, P. W., Baik, S., Heller, D. A., & Strano, M. S. (2005). Near-infrared optical sensors
based on single-walled carbon nanotubes. Nat Mater 4, 8692.
Baron, P. A., Deye, G. J., Chen, B. T., Schwegler-Berry, D. E., Shvedova, A. A., & Castranova, V.
(2008). Aerosolization of single-walled carbon nanotubes for an inhalation study.
Inhal Toxicol 20(8), 751760.
Belo, M. A., Schalch, S. H., Moraes, F. R., Soares, V. E., Otoboni, A. M., & Moraes, J. E.
(2005). Effect of dietary supplementation with vitamin E and stocking density on
macrophage recruitment and giant cell formation in the teleost sh, Piaractus
mesopotamicus. J Comp Pathol 133(23), 146154.
Casey, A., Herzog, E., Davoren, M., Lyng, F. M., Byrne, H. J., & Chambers, G. (2007).
Spectroscopic analysis conrms the interactions between single walled carbon
nanotubes and various dyes commonly used to assess cytotoxicity. Carbon 45,
14251432.
Chakravarty, P., Marches, R., Zimmerman, N. S., Swafford, A. D., Bajaj, P., Musselman, I. H.,
et al. (2008). Thermal ablation of tumor cells with antibody-functionalized singlewalled carbon nanotubes. Proc Natl Acad Sci USA 105, 86978702.
Chen, Z., Meng, H., Xing, G., Chen, C., Zhao, Y., Jia, G., et al. (2006). Acute toxicological
effects of copper nanoparticles in vivo. Toxicol Lett 163, 109120.
Chin, S. F., Baughman, R. H., Dalton, A. B., Dieckmann, G. R., Draper, R. K., Mikoryak, C., et al.
(2007). Amphiphilic helical peptide enhances the uptake of single-walled carbon
nanotubes by living cells. Exp Biol Med (Maywood) 232, 12361244.
Davies, J. C. (2006). Managing the effects of nanotechnology. PEN 2, Woodrow Wilson
International Center for Scholars. Washington, DC.
Davies, J. C. (2008). Nanotechnology oversight: an agenda for the new administration
project on emerging nanotechnologies. PEN 13, Woodrow Wilson International
Center for Scholars. Washington, DC.
Davoren, M., Herzog, E., Casey, A., Cottinerau, B., Chambers, G., Byrne, H. J., et al. (2007).
In vitro toxicity evaluation of single walled carbon nanotubes on human A549 lung
cells. Toxicol in Vitro 21, 438448.
De la Zerda, A., Zavaleta, C., Keren, S., Vaithilingam, S., Bodapati, S., Liu, Z., et al. (2008).
Carbon nanotubes as photoacoustic molecular imaging agents in living mice. Nat
Nanotechnol 3, 557562.
Deng, X., Jia, G., Wang, H., Sun, H., Wang, X., Yang, S., et al. (2007). Translocation and fate
of multi-walled carbon nanotubes in vivo. Carbon 45, 14191424.
Devereux, T. R., Belinsky, S. A., Maronpot, R. R., White, C. M., Hegi, M. E., Patel, A. C., et al.
(1993). Comparison of pulmonary O6-methylguanine DNA adduct levels and Ki-ras
activation in lung tumors from resistant and susceptible mouse strains. Mol
Carcinog 8, 177185.
Donaldson, K. (2006). Resolving the nanoparticles paradox. Nanomedicine 1(2),
229234.

Donaldson, K., Aitken, R., Tran, L., Stone, V., Dufn, R., Forrest, G., et al. (2006). Carbon
nanotubes: a review of their properties in relation to pulmonary toxicology and
workplace safety. Toxicol Sci 92(1), 522.
Donaldson, K., Stone, V., Seaton, A., Tran, L., Aitken, R., & Poland, C. (2008). Re: induction
of mesothelioma in p53 mouse by intraperitoneal application of multi-wall carbon
nanotube. J Toxicol Sci 33, 385388.
Donaldson, K., Tran, L., Jimenez, L. A., Dufn, R., Newby, D. E., Mills, N., et al. (2005).
Combustion-derived nanoparticles: a review of their toxicology following inhalation exposure. Part Fibre Toxicol 2, 10.
Dutta, D., Sundaram, S. K., Teeguarden, J. G., Riley, B. J., Field, L. S., Jacobs, J. M., et al.
(2007). Adsorbed proteins inuence the biological activity and molecular targeting
of nanomaterials. Toxicol Sci 100, 300315.
EPA 40 CFR Part 50 (2006). National Ambient Air Quality Standards or Particulate
Matter; Final Rule volume 71, No. 200.
Fadeel, B., & Kagan, V. E. (2003). Apoptosis and macrophage clearance of neutrophils:
regulation by reactive oxygen species. Redox Rep 8, 143150.
Fadeel, B., Kagan, V., Krug, H., Shvedova, A., Svartengren, M., Tran, L., et al. (2007).
There's plenty of room at the forum: potential risks and safety assessment of
engineered nanomaterials. Nanotoxicology 1(2), 7384.
Fenoglio, I., Greco, G., Tomatis, M., Muller, J., Raymundo-Piero, E., Bguin, F., et al.
(2008). Structural defects play a major role in the acute lung toxicity of multiwall
carbon nanotubes: physicochemical aspects. Chem Res Toxicol 21, 16901697.
Fenoglio, I., Tomatis, M., Lison, D., Muller, J., Fonseca, A., Nagy, J. B., et al. (2006).
Reactivity of carbon nanotubes: free radical generation or scavenging activity? Free
Radic Biol Med 40, 12271233.
Fraire, A. E., & Greenberg, S. D. (1973). Carcinoma and diffuse interstitial brosis of lung.
Cancer 31, 10781086.
Gitlin, J. (2008) Pressure increase in EU to regulate nanotechnology. [updated 2008;
cited]; Available from: http://arstechnica.com/new.ars/post/20080529-pressureincreases-on-eu-to-regulate-nanot
Han, J. H., Lee, E. J., Lee, J. H., So, K. P., Lee, Y. H., Bae, G. N., et al. (2008). Monitoring
multiwalled carbon nanotube exposure in carbon nanotube research facility. Inhal
Toxicol 20, 741749.
Herzog, E., Casey, A., Lyng, F. M., Chambers, G., Byrne, H. J., & Davoren, M. (2007). A new
approach to the toxicity testing of carbon-based nanomaterials. The clonogenic
assay. Toxicol Lett 174, 4960.
Hirsch, L. R., Stafford, R. J., Bankson, J. A., Sershen, S. R., Rivera, B., Price, R. E., et al. (2003).
Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic
resonance guidance. Proc Natl Acad Sci USA 100, 1354913554.
Hubbard, R., Venn, A., Lewis, S., & Britton, J. (2000). Lung cancer and cryptogenic brosing
alveolitis. A population-based cohort study. Am J Respir Crit Care Med 161, 58.
Ichihara, G., Castranova, V., Tanioka, A., & Miyazawa, K. I. (2008). Re: induction of
mesothelioma in p53 mouse by intraperitoneal application of multi-wall carbon
nanotube. J Toxicol Sci 33, 381382 author reply 382.
Iijima, S. (1991). Helical microtubules of graphitic carbon. Nature 354, 5658.
Jacobsen, N. R., Pojana, G., White, P., Mller, P., Cohn, C. A., Korsholm, K. S., et al. (2008).
Genotoxicity, cytotoxicity, and reactive oxygen species induced by single-walled
carbon nanotubes and C(60) fullerenes in the FE1-Mutatrade markMouse lung
epithelial cells. Environ Mol Mutagen 49(6), 476487.
Jia, G., Wang, H., Yan, L., Wang, X., Pei, R., Yan, T., et al. (2005). Cytotoxicity of carbon
nanomaterials: single-wall nanotube, multi-wall nanotube, and fullerene. Environ
Sci Technol 39, 13781383.
Johnson, N. F., Hoover, M. D., Thomassen, D. G., Cheng, Y. S., Dalley, A., & Brooks, A. L.
(1992). In vitro activity of silicon carbide whiskers in comparison to other industrial
bers using four cell culture systems. Am J Ind Med 21(6), 807823.
Kagan, V. E., Bayir, H., & Shvedova, A. A. (2005). Nanomedicine and nanotoxicology: two
sides of the same coin. Nanomedicine 1(4), 313316.
Kagan, V. E., Rao, K. M., Kisin, E. R., Young, S. H., Meighan, T., Murray, A. R., et al. (2007).
Pulmonary effects of single walled carbon nanotubes: inammatory response,
oxidative stress/signaling, and recognition by macrophages. In P. P. Simeonova
(Ed.), NATO Series, Nanotechnology Toxicological Issues and Environmental Safety
(pp. 6575). Springer.
Kagan, V. E., Tyurina, Y. Y., Tyurin, V. A., Konduru, N. V., Potapovich, A. I., Osipov, A. N., et al.
(2006). Direct and indirect effects of single walled carbon nanotubes on RAW 264.7
macrophages: role of iron. Toxicol Lett 165(1), 88100.
Kam, N. W., Liu, Z., & Dai, H. (2006). Carbon nanotubes as intracellular transporters for
proteins and DNA: an investigation of the uptake mechanism and pathway. Angew
Chem Int Ed Engl 45, 577581.
Kam, N. W., O'Connell, M., Wisdom, J. A., & Dai, H. (2005). Carbon nanotubes as
multifunctional biological transporters and near-infrared agents for selective
cancer cell destruction. Proc Natl Acad Sci USA 102, 1160011605.
Karlsson, H. L., Cronholm, P., Gustafsson, J., & Mller, L. (2008). Copper oxide
nanoparticles are highly toxic: a comparison between metal oxide nanoparticles
and carbon nanotubes. Chem Res Toxicol 21(9), 17261732.
Kingston, C. T., & Simard, B. (2006). Recent advances in laser synthesis of single-walled
carbon nanotubes. J Nanosci Nanotechnol 6(5), 12251232.
Kinnula, V. L., & Crapo, J. D. (2004). Superoxide dismutases in malignant cells and
human tumors. Free Radic Biol Med 36(6), 718744.
Kisin, E. R., Murray, A. R., Keane, M. J., Shi, X. C., Schwegler-Berry, D., Gorelik, O., et al.
(2007). Single-walled carbon nanotubes: geno- and cytotoxic effects in lung
broblast V79 cells. J Toxicol Environ Health A 70(24), 20712079.
Knaapen, A. M., Borm, P. J. A., Albrecht, C., & Schins, R. P. F. (2004). Inhaled particles and
lung cancer. Part A: mechanisms. Int J Cancer 109, 799809.
Kostarelos, K., Lacerda, L., Pastorin, G., Wu, W., Wieckowski, S., Luangsivilay, J., et al.
(2007). Cellular uptake of functionalized carbon nanotubes is independent of
functional group and cell type. Nat Nanotechnol 2, 108113.

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204


Kreyling, W. G., Semmler, M., Erbe, F., Mayer, P., Takenaka, S., Schulz, H., et al. (2002).
Translocation of ultrane insoluble iridium particles from lung epithelium to
extrapulmonary organs is size dependent but very low. J Toxicol Environ Health Part
A 65, 15131530.
Kuempel, E., Geraci, C., & Schulte, P. (2007). Risk assessment approaches and research
needs for nanomaterials: an examination of data and information current studies.
In P. Simeonova, N. Oppol, & M. Luster (Eds.), Nanotechnology Toxicological Issues
and Environmental Safety (pp. 119146).
Lacerda, L., Bianco, A., Prato, M., & Kostarelos, K. (2006). Carbon nanotubes as
nanomedicines: from toxicology to pharmacology. Adv Drug Deliv Rev 58, 14601470.
Lam, C. W., James, J. T., McCluskey, R., Arepalli, S., & Hunter, R. L. (2006). A review of
carbon nanotube toxicity and assessment of potential occupational and environmental health risks. Crit Rev Toxicol 36(3), 189217.
Lam, C. W., James, J. T., McCluskey, R., & Hunter, R. L. (2004). Pulmonary toxicity of
single-wall carbon nanotubes in mice 7 and 90 days after intratracheal instillation.
Toxicol Sci 77, 126134.
Li, Z., Hulderman, T., Salmen, R., Chapman, R., Leonard, S. S., Young, S. H., et al. (2007).
Cardiovascular effects of pulmonary exposure to single-wall carbon nanotubes.
Environ Health Perspect 115(3), 377382.
Li, N., Sioutas, C., Cho, A., Schmitz, D., Misra, C., Sempf, J., et al. (2003). Ultrane
particulate pollutants induce oxidative stress and mitochondrial damage. Environ
Health Perspect 111(4), 455460.
Li, N., Xia, T., & Nel, A. E. (2008). The role of oxidative stress in ambient particulate
matter-induced lung diseases and its implications in the toxicity of engineered
nanoparticles. Free Radic Biol Med 44, 16891699.
Lison, D., & Muller, J. (2008). Lung and systemic responses to carbon nanotubes (CNT) in
mice. Toxicol Sci 101, 179180.
Liu, Z., Cai, W., He, L., Nakayama, N., Chen, K., Sun, X., et al. (2007). In vivo biodistribution
and highly efcient tumour targeting of carbon nanotubes in mice. Nat Nanotechnol
2, 4752.
Liu, Z., Davis, C., Cai, W., He, L., Chen, X., & Dai, H. (2008). Circulation and long-term fate
of functionalized, biocompatible single-walled carbon nanotubes in mice probed by
Raman spectroscopy. Proc Natl Acad Sci USA 105, 14101415.
Lloyd's (2007) Risks: nanotechnology (www.lloyds.com/emerging risks).
Magrez, A., Kasas, S., Salicio, V., Pasquier, N., Seo, J. W., Celio, M., et al. (2006). Cellular
toxicity of carbon-based nanomaterials. Nano Lett 6, 11211125.
Maynard, A. D., Aitken, R. J., Butz, T., Colvin, V., Donaldson, K., Oberdrster, G., et al.
(2006). Safe handling of nanotechnology. Nature 444, 267269.
Maynard, A. D., Baron, P. A., Foley, M., Shvedova, A. A., Kisin, E. R., & Castranova, V.
(2004). Exposure to carbon nanotube material: aerosol release during the handling
of unrened single walled carbon nanotube material. J Toxicol Environ Health 67(1),
87107.
Maynard, A. D., & Kuempel, E. D. (2005). Airborne nanostructured particles and
occupational health. J Nanopart Res 7, 587614.
McDevitt, M. R., Chattopadhyay, D., Kappel, B. J., Jaggi, J. S., Schiffman, S. R., Antczak, C.,
et al. (2007). Tumor targeting with antibody-functionalized, radiolabeled carbon
nanotubes. J Nucl Med 48, 11801189.
McDonald, J. & Mitchell, L. (2008) To the Editor. Toxicol. Sci 101: 181182.
Merchant, J. A. (1990). Human epidemiology: a review of ber type and characteristics
in the development of malignant and nonmalignant disease. Environ Health
Perspect 88, 287293.
Mercer, R. R., Scabilloni, J., Wang, L., Kisin, E., Murray, A. R., Schwegler-Berry, D., et al.
(2008). Alteration of deposition pattern and pulmonary response as a result of
improved dispersion of aspirated single-walled carbon nanotubes in a mouse
model. Am J Physiol Lung Cell Mol Physiol 294(1), L8797.
Mitchell, L. A., Gao, J., Wal, R. V., Gigliotti, A., Burchiel, S. W., & McDonald, J. D. (2007).
Pulmonary and systemic immune response to inhaled multiwalled carbon
nanotubes. Toxicol Sci 100, 203214.
Monteiro-Riviere, N. A., & Inman, A. O. (2006). Challenges for assessing carbon
nanomaterial toxicity to skin. Carbon 44, 10701078.
Muller, J., Huaux, F., Fonseca, A., Nagy, J. B., Moreau, N., Delos, M., et al. (2008). Structural
defects play a major role in the acute lung toxicity of multiwall carbon nanotubes:
toxicological aspects. Chem Res Toxicol 21, 16981705.
Muller, J., Huaux, F., Moreau, N., Misson, P., Heilier, J. F., Delos, M., et al. (2005).
Respiratory toxicity of multi-wall carbon nanotubes. Toxicol Appl Pharmacol 207,
221231.
Murr, L. E., Garza, K. M., Soto, K. F., Carrasco, A., Powell, J. G., Ramirez, D. A., et al. (2005).
Cytotoxicity assessment of some carbon nanotubes and related carbon nanoparticle
aggregates and the implication for anthropogenic carbon nanotube aggregates in
the environment. Int J Environ Res Publ Health 2, 3142.
Nel, A., Xia, T., Mdler, L., & Li, N. (2006). Toxic potential of materials at the nanolevel.
Science 311(5761), 622627.
Nemmar, A., Hoet, P. H. M., Vanquickenborne, B., Dinsdale, D., Thomeer, M., Hoylaerts,
M. F., et al. (2002). Passage of inhaled particles into the blood circulation in humans.
Circulation 105, 411414.
Nemmar, A., Vanbilloen, H., Hoylaerts, M. F., Hoet, P. H. M., Verbruggen, A., & Nemery, B.
(2001). Passage of intratracheally instilled ultrane particles from the lung into the
systemic circulation in hamster. Am J Respir Crit Care Med 164, 16651668.
NIOSH (2005). NIOSH current intelligence bulletin: evaluation of health hazard and
recommendations for occupational exposure to titanium dioxide, Unpublished
Public Review Draft, November 22, 2005. US Department of Health and Human
Services, Public Health Service Centers for Disease Control and Prevention, National
Institute for Occupational Safety and Health, Cincinnati, OH; http://www.cdc.gov/
niosh/docs/preprint/tio2/pdfs/TI02Draft./pdf
Oberdrster, G. (1996). Signicance of particle parameters in the evaluation of exposure
doseresponse relationships of inhaled particles. Inhal Toxicol 8(l), 7389.

203

Oberdrster, G. (2007). Biokinetics and Effects of Nanoparticles. In P. P. Simeonova (Ed.),


NATO Series, Nanotechnology Toxicological Issues and Environmental Safety.
Springer, pp 1551.
Oberdrster, G., Ferin, J., Finkelstein, G., Wade, P., & Corson, N. (1990). Increased
pulmonary toxicity of ultrane particles? II. Lung lavage studies. J Aerosol Sci 21(3),
384387.
Oberdrster, G., Ferin, J., & Lehnert, E. (1994). Correlation between particle size, in vivo
particle persistence, and lung injury. Environ Health Perspect 102(5), 173179.
Oberdrster, G., Oberdrster, E., & Oberdrster, J. (2005). Nanotoxicology: an emerging
discipline evolving from studies of ultrane particles. Environ Health Perspect 113(7),
823839.
Oberdrster, G., Sharp, Z., Atudorei, V., Elder, A., Gelein, R., Kreyling, W., et al. (2004).
Translocation of inhaled ultrane particles to the brain. Inhal Toxicol 16, 437445.
Oberdrster, G., Sharp, Z., Atudorei, V., Elder, A., Gelein, R., Lunts, A., et al. (2002).
Extrapulmonary translocation of ultrane carbon particles following whole-body
inhalation exposure of rats. J Toxicol Environ Health Part A 65, 15311543.
Oberdrster, G., Stone, V., & Donaldson, K. (2007). Toxicology of nanoparticles: a
historical perspective. Nanotoxicology 1, 225.
Pacurari, M., Yin, X. J., Ding, M., Leonard, S. S., Schwegler-Berry, D., Ducatman, B. S.,
et al. (2008a). Oxidative and molecular interactions of multi-wall carbon
nanotubes (MWCNT) in normal and malignant human mesothelial cells. Nanotoxicology. 2(3), 155170.
Pacurari, M., Yin, X. J., Zhao, J., Ding, M., Leonard, S. S., Schwegler-Berry, et al. (2008b).
Raw single-wall carbon nanotubes induced oxidative stress and activate MAPKs,
AP-1, NF-Kb, and aKT in normal and malignant human mesothelial cells. Environ
Health Prosp 116(9), 12111217.
Pantarotto, D., Partidos, C. D., Hoebeke, J., Brown, F., Kramer, E., Briand, J. P., et al.
(2003). Immunization with peptide-functionalized carbon nanotubes enhances
virus-specic neutralizing antibody responses. Chem Biol 10, 961966.
Pantarotto, D., Singh, R., McCarthy, D., Erhardt, M., Briand, J. P., Prato, M., et al. (2004).
Functionalized carbon nanotubes for plasmid DNA gene delivery. Angew Chem Int
Ed Engl 43, 52425246.
Poland, C. A., Dufn, R., Kinloch, I., Maynard, A., Wallace, W. A., Seaton, A., et al.
(2008). Carbon nanotubes introduced into the abdominal cavity of mice show
asbestos-like pathogenicity in a pilot study. Nat Nanotechnol 3(7), 423428.
Pope, C. A., III (2000). Epidemiology of ne particulate air pollution and human health:
biologic mechanisms and who's at risk? Environ Health Perspect 108(Suppl 4),
713723.
Porter, D. W., Sriram, K., Wolfarth, M. G., Jefferson, A. M., Schwegler-Berry, D.,
Andrew, M. E., & Castranova, V. (2008). A biocompatible medium for nanoparticle
dispersion. Nanotoxicology 2, 144154.
Prato, M., Kostarelos, K., & Bianco, A. (2008). Functionalized carbon nanotubes in drug
design and discovery. Acc Chem Res 41, 6068.
Pritchard, J. B., French, J. E., Davis, B. J., & Haseman, J. K. (2003). The role of transgenic
mouse models in carcinogen identication. Environ Health Perspect 111(4), 444454.
Przybyla, D. E., & Chmielewski, J. (2008). Metal-triggered radial self-assembly of
collagen peptide bers. J Am Chem Soc 130, 1261012611.
Pulskump, K., Drabate, S., & Krug, H. F. (2007). Carbon nanotubes show no sign of acute
toxicity but induce intracellular reactive oxygen species in dependence on
contaminants. Toxicol Lett 168, 5874.
Rocksn, D., Ekstrand-Hammarstrm, B., Johansson, L., & Bucht, A. (2003). Vitamin E
reduces transendothelial migration of neutrophils and prevents lung injury in
endotoxin-induced airway inammation. Am J Respir Cell Mol Biol 28(2), 199207.
Roco, M. C. (2006). Progress in governance of converging technologies integrated from
the nanoscale. Ann N Y Acad Sci 1093, 123.
Ronai, Z. A., Gradia, S., Peterson, L. A., & Hecht, S. S. (1993). G to A transitions and G to T
transversions in codon 12 of the Ki-ras oncogene isolated from mouse lung tumors
induced by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and related
DNA methylating and pyridyloxobutylating agents. Carcinogenesis 14, 24192422.
Sadowska, A. M., Verbraecken, J., Darquennes, K., & De Backer, W. A. (2006). Role of Nacetylcysteine in the management of COPD. Int J Chron Obstruct Pulmon Dis 1(4),
425434.
Sager, T., Porter, D., Robinson, V., Lindsley, W. G., Schwegler-Berry, D. E., & Castranova,
V. (2007). An improved method to disperse nanoparticles for in vitro and in vivo
investigation of toxicity. Nanotoxicol 1, 118129.
Salvemini, D., Muscoli, C., Riley, D. P., & Cuzzocrea, S. (2002). Superoxide dismutase
mimetics. Pulm Pharmacol Ther 15(5), 439447.
Sayes, C. M., Liang, F., Hudson, J. L., Mendez, J., Guo, W., Beach, J. M., et al. (2006).
Functionalization density dependence of single-walled carbon nanotubes cytotoxicity in vitro. Toxicol Lett 161(2), 135142.
Seluk, Z. T., Cpl, L., Emri, S., Kalyoncu, A. F., Sahin, A. A., & Bari, Y. I. (1992).
Malignant pleural mesothelioma due to environmental mineral ber exposure in
Turkey. Analysis of 135 cases. Chest 102(3), 790796.
Singh, U., Devaraj, S., & Jialal, I. (2005). Vitamin E, oxidative stress, and inammation.
Annu Rev Nutr 25, 151174.
Singh, S., & Nalwa, H. S. (2007). Nanotechnology and health safetytoxicity and risk
assessments of nanostructured materials on human health. J Nanosci Nanotechnol 7(9),
30483070.
Singh, R., Pantarotto, D., Lacerda, L., Pastorin, G., Klumpp, C., Prato, M., et al. (2006).
Tissue biodistribution and blood clearance rates of intravenously administered
carbon nanotube radiotracers. Proc Natl Acad Sci USA 103, 33573362.
Sint, T., Donohue, J. F., & Ghio, A. J. (2008). Ambient air pollution particles and the acute
exacerbation of chronic obstructive pulmonary disease. Inhal Toxicol 20(1), 2529.
Shvedova, A. A., Fabisiak, J. P., Kisin, E. R., Murray, A. R., Roberts, J. R., Tyurina, Y. Y., et al.
(2008a). Sequential exposure to carbon nanotubes and bacteria enhances pulmonary
inammation and infectivity. Am J Respir Cell Mol Biol 38(5), 579590.

204

A.A. Shvedova et al. / Pharmacology & Therapeutics 121 (2009) 192204

Shvedova, A. A., Kisin, E. R., Mercer, R., Murray, A. R., Johnson, V. J., Potapovich, A. I., et al.
(2005). Unusual inammatory and brogenic pulmonary responses to single-walled
carbon nanotubes in mice. Am J Physiol 289, L698L708.
Shvedova, A. A., Kisin, E. R., Murray, A. R., Gorelik, O., Arepalli, S., Castranova, V., et al.
(2007a). Vitamin E deciency enhances pulmonary inammatory response and
oxidative stress induced by single-walled carbon nanotubes in C57BL/6 mice. Toxicol Appl Pharmacol 221(3), 339348.
Shvedova, A. A., Kisin, E., Murray, A. R., Johnson, V. J., Gorelik, O., Arepalli, S., et al.
(2008b). Inhalation versus aspiration of single walled carbon nanotubes in C57BL/6
mice: inammation, brosis, oxidative stress and mutagenesis. Am J Physiol Lung
Cell Mol Physiol 295, 552565.
Shvedova, A. A., Kisin, E. R., Murray, A. R., Kommineni, C., Castranova, V., Fadeel, B., et al.
(2008c). Increased accumulation of neutrophils and decreased brosis in the lung of
NADPH oxidase-decient C57BL/6 mice exposed to carbon nanotubes. Toxicol Appl
Pharmacol 231, 235240.
Shvedova, A. A., Kisin, E. R., Murray, A. R., Schwegler-Berry, D., Gandelsman, V. Z.,
Baron, P., et al. (2004). Exposure of human bronchial epithelial cells to carbon
nanotubes caused oxidative stress and cytotoxicity. Proceedings of the Society for
Free Radical Research Meeting, European Section, June 2629, 2003, Ioannina, Greece
(pp. 91103).
Shvedova, A. A., Sager, T., Murray, A., Kisin, E., Porter, D. W., Leonard, S. S., et al.
(2007b). Critical issues in the evaluation of possible effects resulting from airborne
nanoparticles. In N. Monteiro-Riviere & L. Train (Eds.), Nanotechnology: Characterization, Dosing and Health Effects (pp. 221232). Philadelphia: Informa Healthcare
Chap 14.
Soto, K. F., Carrasco, A., Powell, T. G., Garza, K. M., & Murr, L. E. (2005). Comparative in
vitro cytotoxicity assessment of some manufactured nanoparticulate materials
characterized by transmission microscopy. J Nanopart Res 7, 145169.
Soto, K., Garza, K. M., & Murr, L. E. (2007). Cytotoxic effects of nanomaterials. Acta
Biomaterialia 3, 351358.
Sriram, K., Porter, D., Tsuruoka, S., Endo, M., Jefferson, A., Wolfarth, W., et al. (2007).
Neuroinammatory responses following exposure to engineered nanomaterials.
Toxicologist A1390.
Stern, S. T., & McNeil, S. E. (2008). Nanotechnology safety concerns revisited. Toxicol Sci
101, 421.
Szendi, K., & Varga, C. (2008). Lack of genotoxicity of carbon nanotubes in a pilot study.
Anticancer Res 28(1A), 349352.
Takagi, A., Hirose, A., Nishimura, T., Fukumori, N., Ogata, A., Ohashi, N., et al. (2008).
Induction of mesothelioma in p53+/ mouse by intraperitoneal application of
multi-wall carbon nanotube. J Toxicol Sci 33(1), 105116.
Vaslet, C. A., Messier, N. J., & Kane, A. B. (2002). Accelerated progression of asbestosinduced mesotheliomas in heterozygous p53+/ mice. Toxicol Sci 68, 331338.

Vinzents, P. S., Mller, P., Srensen, M., Knudsen, L. E., Hertel, O., Jensen, F. P., et al.
(2005). Personal exposure to ultrane particles and oxidative DNA damage. Environ
Health Perspect 113(11), 14851490.
Walker, & Mouton (2008). Environmental inuences on cardiovascular health. J Natl
Med Assoc 100(1), 98102.
Wang, L., Castranova, V., Rojanasakul, Y., Lu, Y., Scabilloni, J., & Mercer, R. R. (2008).
Direct brogenic effects of dispersed single walled carbon nanotubes on human
lung broblasts. Toxicologists 102, A1499.
Wang, H., Wang, J., Deng, X., Sun, H., Shi, Z., Gu, Z., et al. (2004). Biodistribution of
carbon single-wall carbon nanotubes in mice. J Nanosci Nanotechnol 4, 10191024.
Warheit, D. B., Laurence, B. R., Reed, K. L., Roach, D. H., Reynolds, G. A., & Webb, T. R.
(2003). Comparative pulmonary toxicity assessment of single wall carbon
nanotubes in rats. Toxicol Sci 26, 22812300.
Wick, P., Manser, P., Limbach, L. K., Dettlaff-Weglikowska, U., Krumeich, F., Rothet, S., et al.
(2007). The degree and kind of agglomeration affect carbon nanotube toxicity. Toxicol
Lett 168, 121131.
Wick, P., Manser, P., Spohn, P., & Bruinink, A. (2006). In vitro evaluation of possible
adverse effects of nanosized materials. Phys Status Solidi 243, 35563560.
Witasp, E., Kagan, V., & Fadeel, B. (2008). Programmed cell clearance: molecular
mechanisms and role in autoimmune disease, chronic inammation, and anticancer immune responses. Curr Immunol Rev 4, 5369.
Worle-Knirsch, J. M., Pulskamp, K., & Krug, H. F. (2006). Oops they did it again! Carbon
nanotubes hoax scientists in viability assays. Nano Lett 6, 12611268.
Wright, C. E., Tallan, H. H., Lin, Y. Y., & Gaull, G. E. (1986). Taurine: biological update. In C.
C. Richardson, P. D. Boyer, I. B. Dawid, & A. Meister (Eds.), Annual Review of
Biochemistry Palo Alto: Annual Reviews, Inc.
Yacobi, N. R., Phuleria, H. C., Demaio, L., Liang, C. H., Peng, C. A., Sioutas, C., et al.
(2007). Nanoparticle effects on rat alveolar epithelial cell monolayer barrier
properties. Toxicol in Vitro 21, 13731381.
Yang, S., Guo, W., Lin, Y., Deng, X., Wang, H., Sun, H., et al. (2007). Biodistribution of
pristine single-walled carbon nanotubes in vivo. J Phys Chem C 111, 1776117764.
Zhang, Q., Kusaka, Y., & Donaldson, K. (2000). Comparative pulmonary responses
caused by exposure to standard cobalt and ultrane cobalt. J Occup Health 42,
179184.
Zhang, Q., Kusaka, Y., Zhu, X., Sato, K., Mo, Y., Klutz, T., et al. (2003). Comparative
toxicity of standard nickel and ultrane nickel in lung after intratracheal
instillation. J Occup Health 45, 2330.
Zhang, L. W., Zeng, L., Barron, A. R., & Monteiro-Riviere, N. A. (2007). Biological
interactions of functionalized single-wall carbon nanotubes in human epidermal
keratinocytes. Int J Toxicol 26(2), 103113.
Zhu, L., Chang, D. W., Dai, L., & Hong, Y. (2007). DNA damage induced by multiwalled
carbon nanotubes in mouse embryonic stem cells. Nano Lett 7(12), 35923597.

You might also like