ArticlePDF Available

What are carbon nanotubes’ roles in anti-tumor therapies?

Authors:
  • Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College

Abstract

Since their discovery, carbon nanotubes (CNTs) have become one of the most promising nanomaterials in many industrial and biomedical applications. Due to their unique physicochemical properties, CNTs have been proposed and actively exploited as multipurpose innovative carriers for cancer therapy. The aim of this article is to provide an overview of the status of applications, advantages, and up-to-date research and development of carbon nanotubes in cancer therapy with an emphasis on drug delivery, photothermal therapy, gene therapy, RNAi, and immune therapy. In addition, the issues of risk and safety of CNTs in cancer nanotechnology are discussed briefly. Keywordscarbon nanotubes-tumor-therapy-delivery-immune
SCIENCE CHINA
Chemistry
© Science China Press and Springer-Verlag Berlin Heidelberg 2010 chem.scichina.com www.springerlink.com
*Corresponding author (email: xuhy@pumc.edu.cn)
ARTICLES November 2010 Vol.53 No.11: 1–7
doi: 10.1007/s11426-010-4117-6
What are carbon nanotubes’ roles in anti-tumor therapies?
XU HaiYan*, MENG Jie & KONG Hua
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
Received May 12, 2010; accepted July 4, 2010
Since their discovery, carbon nanotubes (CNTs) have become one of the most promising nanomaterials in many industrial and
biomedical applications. Due to their unique physicochemical properties, CNTs have been proposed and actively exploited as
multipurpose innovative carriers for cancer therapy. The aim of this article is to provide an overview of the status of applica-
tions, advantages, and up-to-date research and development of carbon nanotubes in cancer therapy with an emphasis on drug
delivery, photothermal therapy, gene therapy, RNAi, and immune therapy. In addition, the issues of risk and safety of CNTs in
cancer nanotechnology are discussed briefly.
carbon nanotubes, tumor, therapy, delivery, immune
1 Introduction
In the continuous fight against cancer, there are many chal-
lenges that reduce the efficiency of existing antitumor
therapies such as serious side effects of chemotherapy, mul-
tidrug resistance (MDR) and immune escape mechanisms of
tumor cells. In recent years, rapid advances in nanoparti-
cle-based delivery systems have provided promising new
ways to overcome those challenges. Among the various
nanoparticles, carbon nanotubes (CNTs) have recently gar-
nered intense research interest and have shown attractive
performance in various aspects of antitumor therapies.
CNTs have a unique structure on the nanoscale that al-
lows them to enter cells easily and to conjugate various
therapeutic molecules. Additionally, their pure carbon
composition makes them possess good biocompatibility. In
light of these factors, CNTs have become a new class of
promising candidates for a delivery system. This review
attempts to provide insight into the current research pro-
gress using CNTs for cancer therapy.
2 The role of CNTs in chemotherapy
Chemotherapy is currently one major strategy to fight can-
cers. However, patients have to be administered a combina-
tion of drugs that increase in dose over time, which results
in severe side effects, including toxicity and multidrug re-
sistance (MDR). Hence, development of new delivery vehi-
cles to transport chemotherapeutic reagents into tumor cells
with high efficiency is necessary to overcome drug resis-
tance as well as to reduce side effects. An emerging ap-
proach is to utilize nanotechnology to optimize the existing
chemotherapies. Carbon nanotube-based drug delivery has
shown a promise in tumor-targeted accumulation in mice,
and the system exhibits good biocompatibility, good excre-
tability, and little toxicity. Various chemotherapeutic re-
agents have been demonstrated to have increased efficiency
after being chemically linked to CNTs.
2.1 Delivery of chemotherapeutic drugs by carbon
nanotubes
Paclitaxel (PTX) is one of the most widely used cancer
chemotherapy drugs. When PTX was conjugated to branched
2 XU HaiYan, et al. Sci China Chem November (2010) Vol.53 No.11
polyethylene glycol chains on single-walled carbon nano-
tubes (SWNTs) via a cleavable ester bond, the SWNT-PTX
affords higher efficacy in suppressing tumor growth than
clinical Taxol in a murine 4T1 breast cancer model. The
improvement is owing to the prolonged circulation in the
blood and a 10-fold higher PTX uptake by the tumor when
delivered by SWNTs (likely because of an enhanced per-
meability and retention (EPR) effect). Drug molecules car-
ried into the reticuloendothelial system are released from
SWNTs and excreted via the biliary pathway without caus-
ing apparent toxic effects to normal organs [1]. Multiwalled
carbon nanotubes (MWNTs) perform similarly when carry-
ing chemotherapeutic reagents into tumor cells. When anti-
tumor reagent 10-hydroxycamptothecin (HCPT) was linked
to MWNTs covalently through a cleavable ester linkage
using hydrophilic diaminotriethylene glycol as the spacer
between the nanotube and drug moieties, the obtained
MWNT- HCPT conjugates were found to be superior to the
clinical HCPT formulation in antitumor activity both in vi-
tro and in vivo [2]. The MWNT-HCPT conjugates also have
relatively long circulation in the blood and high drug accu-
mulation at the tumor site. These properties together with
the enhanced cellular uptake and the multivalent presenta-
tion of HCPT on a single nanotube substantially benefit the
antitumor effects.
It is noteworthy that the cytotoxicity of chemotherapeutic
reagents has been demonstrated to be associated with the
linkers that combine drug molecules to CNTs. When meth-
otrexate was tethered to MWNTs through different cleav-
able linkers, for example, the conjugates were internalized
by human breast cancer cells, the initial investigations using
such conjugates indicated that the activity of the drug
strongly depends on the type of the linker used. High cyto-
toxic activity could be observed when using an enzyme-
sensitive peptide linker [3]. For example, single-walled
carbon nanotubes (SWNTs) were conjugated to a doxorubi-
cin prodrug via a carbamate linker that cleaves enzymati-
cally to allow temporal release of the active drug. The con-
jugate was rapidly internalized into the lysosomes of mela-
noma cells and was retained in the subcellular compartment
for over 24 h. The conjugate induced time-dependent cell
death in B16-F10 melanoma cells in vitro and inhibited
melanoma tumors in vivo without the systemic side effects
associated with free doxorubicin [4].
Physical combination is another effective way to deliver
chemotherapeutic reagents. It is theorized that drug mole-
cules can physically conjugate with CNTs via pi-pi stacking
to form supramolecular complexes with the aromatic chro-
mophore and enhance their cytotoxic activity [5, 6]. Immer-
sion of poly (ethylene glycol)-graft-SWNTs (PEG-g-SWNTs)
or PEG-graft-MWNTs (PEG-g-MWNTs) in a saturated so-
lution of PTX in methanol can achieve physical loading of
PTX onto CNTs. Both PTX-loaded PEG-g-SWNTs and
PTX-loaded PEG-g-MWNTs are highly efficient in killing
HeLa and MCF-7 cells [7]. PEG-doxorubicin conjugate
(PEG-DXR) was adsorbed onto oxidized single-wall carbon
nanohorns (oxSWNHs). The intratumorally administered
PEG-DXR-bound oxSWNHs caused a significant retarda-
tion in tumor growth associated with the prolonged DXR
retention in the tumor. In accordance with this DXR reten-
tion, a large number of oxSWNH agglomerates were found
from the periphery of the tumor to the axillary lymph node,
which is a major site of breast cancer metastasis near the
tumor, possibly by means of interstitial lymphatic-fluid
transport [8].
Another strategy to load chemotherapeutic reagents
physically is to fill the drugs into the tubes. For example,
CNTs can carry the antitumor drugs cisplatin and car-
boplatin inside by using a nanoprecipitation method or a
wet-chemical approach after the CNTs were opened [9, 10].
The conjugates were observed adhering to the cell surfaces
in vitro and stayed within the tumor tissues in vivo, which
implies that the drugs were released from the CNTs in high
concentrations locally at the cells in vitro and in the tissues
in vivo and could efficiently attack the tumor cells. When
intratumorally injected into the transplanted tumors of mice,
the conjugate suppressed the tumor growth more than intact
cisplatin. However, compared with the chemical binding
and physical adsorption methods mentioned above, filling
drugs into CNTs has not been widely employed partially
because the preparation is intricate and the release mecha-
nism from the CNTs is still unknown. It is also noted that in
some studies researchers found that CNTs themselves had
an in vivo anticancer effect in addition to significantly en-
hancing the chemotherapeutic effects of the drugs [9, 11].
2.2 Tumor-targeting carbon nanotubes in chemotherapy
A long-standing problem in cancer chemotherapy is the lack
of tumor-specific treatments. Cytotoxic reagents have very
little or no specificity, which leads to systemic toxicity and
causes undesirable severe side effects. CNTs passively re-
alize their targeting function due to the EPR effect of the
nanoparticles; therefore, specificity is still one main obsta-
cle in targeting the delivery of antitumor drugs. Addition-
ally, the lack of tumor-specific markers stands as another
big challenge. Therefore, the development of innovative and
efficacious tumor-specific drug delivery protocols or sys-
tems is urgently needed to fight against various cancer types,
especially multidrug-resistant tumors.
Usually tumor cells overexpress some tumor associated
antigens (TAA) and/or tumor specific antigens (TSA) that
can be used as targets to deliver cytotoxic agents into tu-
mors. Accordingly, the types of targeting molecules mainly
include peptides, proteins, and small chemical molecules
such as folate. The transporting capabilities of CNTs com-
bined with antibodies that recognize tumor-associated pro-
teins or receptors may further improve the efficiency and
reduce the toxic side effects of chemotherapeutic reagents.
One example of a targeting protein is HER2. Recom-
XU HaiYan, et al. Sci China Chem November (2010) Vol.53 No.11 3
bined ricin A chain protein (RTA) is a toxin protein that
induces cell death. When coupled with HER2, the delivery
of RTA via nanotube carriers can selectively recognize the
HER2/neu receptor on certain breast cancer cells and induce
targeted destruction of the tumor cells [12]. Receptors of
various growth factors are other possible protein targets on
the tumor cells. When cisplatin and epidermal growth fac-
tors (EGF) were attached to SWNTs to specifically target
squamous cancer, the SWCNT-cisplatin-EGF was selec-
tively taken up by the HNSCC tumors of mice. Most sig-
nificantly, regression of tumor growth was rapid in mice
treated with targeted SWNT-cisplatin-EGF relative to non-
targeted SWNT-cisplatin [13].
Folic acid (FA) is a targeting molecule for many tumors
as the folate receptor (FR) is overexpressed across a broad
spectrum of human tumors. Most low-molecular-weight
platinum anticancer drugs have short blood circulation
times. Researchers chemically conjugated a platinum (IV)
complex containing a folate derivative at an axial position
to the surface of amine-functionalized SWNTs. The SWNTs
deliver the folate-bearing Pt (IV) cargos into FR (+) cancer
cells by endocytosis. Once inside the cell, cisplatin, formed
upon reductive release from the SWNTs, enters the nucleus
and reacts with the nuclear DNA to which it is targeted [14].
A combination of FA modification with magnetic CNTs
may facilitate the targeted delivery of drugs in the lym-
phatic tissue more effectively. Chemotherapeutic agents
were incorporated into the pores of functionalized magnetic
CNTs, and FA was non-covalently conjugated to the mag-
netic CNTs. By using an externally placed magnet to guide
the drug matrix to the region of the targeted lymph nodes,
the magnetic CNTs can be retained in the draining lymph
nodes for several days while continuously releasing chemo-
therapeutic drugs [15]. FA coupling can be combined with
pH sensitive modifications in CNTs. Polysaccharide coating
and FA modification of SWNTs form a dual-targeting de-
livery system that can selectively deliver DOX into HeLa
cells and release the drug in the lysosomes at low pH with a
much higher efficiency to damage the nuclear DNA and
inhibit cell proliferation [16].
2.3 Using carbon nanotubes to overcome multidrug
resistance
Multidrug resistance (MDR), which is related to cancer
chemotherapy, tumor stem cells, and tumor metastasis, is a
huge obstacle for effective cancer therapy. One of the un-
derlying mechanisms of MDR is the increased efflux of
anticancer drugs by overexpressed P-glycoprotein (P-gp) in
multidrug resistant cells. To overcome MDR in K562 hu-
man leukemia cells, functionalized water-soluble SWNTs
conjugated to the antibody of P-gp (Ap) and loaded with
doxorubicin (Dox), or Dox/Ap-SWNTs, were synthesized.
The binding affinity of Ap-SWNTs toward drug-resistant
K562R cells was 23-fold higher than that toward drug-
sensitive K562S cells. Additionally, Ap-SWNTs could spe-
cifically localize on the cell membrane of K562R cells, and
the amount of Dox in K562R cells could be significantly
enhanced when Ap-SWNTs were employed as a carrier.
Moreover, the Dox/Ap-SWNTs expressed 2.4-fold higher
cytotoxicity and showed a significant suppression of cell
proliferation for K562R leukemia cells as compared to free
Dox [17].
3 CNTs help physical treatments to kill tumor cells
3.1 CNTs’ synergic role with NIR irradiation
Photothermal therapy using nanomaterials has recently at-
tracted interest as an efficient strategy for the next genera-
tion of cancer treatments. Hyperthermia therapy is one op-
tional solution for cancer treatment especially for some
kinds of solid malignant tumors such as liver and breast
tumors. CNTs are unique materials that absorb infrared ra-
diation (IR) especially between 700 and 1100 nm where
body tissues, including skin, are most transparent. This in-
trinsic property of CNTs can be used to optically stimulate
nanotubes located inside living cells to afford multifunc-
tional nanotube biological transporters.
SWNTs are an up-and-coming, potent candidate to be a
photothermal therapeutic agent because they generate sig-
nificant amounts of heat upon excitation with near-infrared
light. Continuous NIR radiation can cause cell death be-
cause of excessive local heating of SWNTs. Such a photo-
thermal effect can be employed to induce thermal cell death
in a noninvasive manner. Combined treatments using SWNTs
and NIR irradiation completely destroyed the tumors with-
out harmful side effects or recurrence of the tumors over six
months in the tumor-loaded mice while the tumors treated
in other control groups grew continuously until the death of
the mice. Most of the injected SWNTs were almost com-
pletely excreted from the bodies of the mice in about 2
months through either the biliary or urinary pathway [18].
MWNTs also strongly absorb near-infrared radiation and
efficiently convert the absorbed energy into released heat.
MWNTs can be stimulated with NIR irradiation to destroy
Erlich ascitic carcinoma (EAC) cells [19] and cervical can-
cer HeLa cells [20]. It was demonstrated that the addition of
MWNTs to a suspension of tumor cells results in the photo-
ablative destruction of cells exposed to short time NIR irra-
diation. The use of MWNTs to generate NIR-induced heat
also led to the thermal destruction of kidney cancers in vitro
and in vivo with minimal local toxicity and no evident sys-
temic toxicity, which was demonstrated through magnetic
resonance temperature-mapping and heat shock protein-
reactive immunohistochemistry [21, 22].
3.2 Drug-loading or targeting carbon nanotubes in
cancer photothermal therapy
A combination of the chemotherapeutic agents oxaliplatin
4 XU HaiYan, et al. Sci China Chem November (2010) Vol.53 No.11
or mitomycin C and rapid heating stimulated by IR has
shown effectiveness in killing colorectal cancer cells. This
approach may be used as a rapid bench-to-bedside clinical
therapeutic agent with a significant impact in the localiza-
tion of chemotherapy agents during the surgical manage-
ment of peritoneal dissemination of colorectal cancer [23].
Filling CNTs with magnetic materials offers the potential
for hyperthermia applications [24].
Specificity is also necessary for CNTs used in the ther-
mal ablation of cancers. The transporting capabilities of
CNTs combined with suitable functionalization chemistry
and their intrinsic optical properties can lead to new classes
of novel nanomaterials for the photothermal therapy of
cancer. CNTs attached to antibodies or peptides represent a
typical approach to targeting cancer cells. For example, to
target disialoganglioside (GD2) specifically, which is over-
expressed on the surface of neuroblastoma stNB-V1 cells,
GD2 monoclonal antibody (anti-GD2) was conjugated to
acidified CNTs. The anti-GD2-linked CNTs were exten-
sively internalized by neuroblastoma cells via GD2-mediated
endocytosis. In addition, the anti-GD2 bound CNTs were
not ingested by PC12 cells, which had no GD2 expression.
After NIR laser exposure, stNB-V1 cells were all found to
undergo necrosis while non-GD2 expressing PC12 cells all
remained viable [25]. CD22, which is broadly expressed on
human B cell lymphomas, and HER2, which is usually
overexpressed on various tumor cells, are other representa-
tive proteins that can be targeted by CNTs [26–28].
Conjugated with folate and irradiated by a 980-nm laser
[29] or a 1064-nm Q-switched millisecond pulsed laser [30],
the modified SWNTs effectively enhanced the photothermal
destruction of tumor cells and noticeably spared nontargeted
normal cells. Selective cancer cell destruction can be
achieved by functionalization of SWNTs with a folate moi-
ety followed by selective internalization of SWNTs inside
cells labeled with folate receptor tumor markers and NIR-
triggered cell death without harming receptor-free normal
cells [31]. DNA-encasement has been reported to increase
heat emission following NIR irradiation of MWNTs, and
DNA-encased MWNTs can be used to safely eradicate a
tumor mass in vivo. Upon irradiation, DNA-encasement
resulted in a 3-fold reduction in the concentration of MWNTs
required to impart a 10 °C temperature increase in bulk solu-
tion. A single treatment consisting of intratumoral injection
of MWNTs followed by 2.5 W/cm2 of laser irradiation at 1064
nm completely eradicated PC3 xenograft tumors in 100%
(8/8) of nude mice. Tumors that received only the MWNTs
injection or the laser irradiation showed growth rates indis-
tinguishable from untreated control tumors. Nonmalignant
tissues displayed no long-term damage from treatment [32].
3.3 Carbon nanotubes enhance the efficacy of other
physical cancer treatments
SWNTs have shown their ability to release heat when
placed in a radiofrequency (RF) field, which has been used
to produce thermal cytotoxicity in three human cancer cell
lines. The RF field induced the efficient heating of aqueous
suspensions of SWNTs. Direct intratumoral injection of
SWNTs in vivo followed by immediate RF field treatment
was tolerated well by rabbits bearing hepatic VX2 tumors.
At 48 h, all SWCNT-treated tumors demonstrated complete
necrosis whereas control tumors that were treated with RF
but without SWNTs remained completely viable. Tumors
that were injected with SWNTs but not treated with RF
were also viable [33].
SWNTs have also shown the potential to enhance dielec-
tric contrast between malignant and normal tissues during
microwave detection of breast cancer, and they facilitate
selective heating of malignant tissue for microwave hyper-
thermia treatment of breast cancer. At SWNT concentra-
tions of less than 0.5% by weight, significant increases in
the relative permittivity and effective conductivity could be
observed, which led to a significantly greater temperature
increase in microwave heating experiments. This work is a
first step toward the development of functionalized, tu-
mor-targeting SWNTs as integrated therapeutic and diag-
nostic agents for microwave breast cancer detection and
treatment [34].
4 Carbon nanotubes in antitumor gene therapy
Gene therapy and RNAi have presented great potential for
antitumor treatment. However, gene therapy relies on the
efficient and nontoxic transport of therapeutic genetic
medicine through the cell membranes. Recent studies have
reported that CNTs can deliver a large amount of therapeu-
tic agents, including DNA and RNA, to the target disease
sites. In addition, due to their unparalleled optical and elec-
trical properties, CNTs can deliver DNA/siRNA into cells
including some difficult-to-transfect primary-immune cells
and bacteria; they can also lead to controlled release of
DNA/siRNA for targeted gene therapy. Furthermore, due to
their wire shaped structure (with a diameter matching that
of DNA/siRNA) and their remarkable flexibility, CNTs can
influence the conformational structure and the transient
conformational changes of DNA/RNA, which can further
enhance the therapeutic effects of DNA/siRNA. Synergistic
combinations of the CNT’s capabilities to deliver DNA/
siRNAs will lead to the development of powerful multi-
functional nanomedicine to treat cancer [35].
The ability to obtain a biological response to SWNT/
siRNA complexes has been seen in a variety of cancer cell
types. By noncovalently conjugating unmodified siRNA with
pristine SWNTs, the conjugates containing siRNA targeted
to hypoxia-inducible factor 1 alpha (HIF-1alpha) showed
strong specific inhibition of cellular HIF-1alpha activity.
Moreover, intratumoral administration of SWCNT-HIF-
1alpha siRNA complexes in mice bearing MiaPaCa-2/HRE
XU HaiYan, et al. Sci China Chem November (2010) Vol.53 No.11 5
tumors significantly inhibited the activity of tumor HIF-
1alpha [36]. Another example is the treatment of a human
lung carcinoma model in vivo using siRNA sequences,
which led to cytotoxicity and cell death using amino-func-
tionalized multiwalled carbon nanotubes (MWNT-NH3
+).
The MWNT-NH3
+:siRNA complexes administered intratu-
morally can elicit delayed tumor growth and increase sur-
vival of xenograft-bearing animals, which is believed to
activate biologically in vivo by triggering an apoptotic cas-
cade that leads to extensive necrosis of the human tumor
mass followed by a concomitant prolongation of survival of
human lung tumor-bearing animals [37].
Cyclin A(2) plays a critical role in DNA replication,
transcription, and cell cycle regulation. It is overexpressed
by many types of cancers including leukemia, which sug-
gests that suppression of cyclin A(2) would be an attractive
strategy to prevent tumor progression. Research results
show that functionalized SWNTs can facilitate the coupling
of siRNA that specifically targets human cyclin A(2) to
form cyclin A(2) siRNA-f-SWNTs complexes. These func-
tionalized SWNTs readily enter K562 cells resulting in the
suppression of cyclin A(2) expression. This provides new
insights into additional therapeutic options for chronic
myelogenous leukemia beyond chemotherapy in light of
increasing multidrug resistance [38]. With the aim of im-
proving the amount and delivery efficiency of genes carried
by carbon nanotubes into human cancer cells, different gen-
erations of polyamidoamine dendrimers were used to mod-
ify MWNTs. The modified CNTs can be fully conjugated
with antisense c-myc oligonucleotides (asODN). The resul-
tant conjugate of asODN-MWNTs entered tumor cells
within 15 min (including human breast cancer MCF-7 cells,
MDA-MB-435 cells, and liver cancer HepG2 cells), inhib-
ited the cells’ growth in a time- and dose-dependent manner,
and down-regulated the expression of the c-myc gene and
C-Myc protein [39].
5 CNTs’ role in cancer immune therapy
The significance of immunotherapy as an adjuvant anti-
cancer treatment has been well recognized [40–42]. While
chemotherapy faces the issues of accumulative toxicity and
drug resistance, anti-tumor immunotherapy usually has few
adverse effects, good patient tolerance, and the potential to
improve the prognosis significantly [43]. Some clinical tri-
als of immunotherapy achieved promising results in treating
malignancies such as melanoma, malignant glioma, or renal
cell carcinoma, which tended to respond poorly to chemo-
therapies [44–47]. However, the efficacy of current immu-
notherapy generally needs further improvement.
Tumor development and anti-tumor mechanisms of the
host are critical issues with large complexity and big chal-
lenges. Cancer cells often develop immune tolerance and
immune escape mechanisms [48, 49]; therefore, the anti-
tumor activities of the immune cells in tumor-bearing hosts
are at a very low level, which allows most tumor cells to
escape the immune system [50]. To mount an anti-cancer
immune reaction, one area of particular interest is CNTs’
role in modulating immunological functions. It has been
reported that functionalized CNTs are non-cytotoxic to im-
mune cells [51]. In previous work, we have investigated the
immunological responses that MWNTs may induce in a
tumor-bearing mice model via a local administration of
subcutaneous injection, and we explored the possibility of
utilizing the immune responses to modify the progress of
tumor progression. We have demonstrated that the subcuta-
neously injected water-soluble MWNTs (wsMWNT) can
act as an immunostimulatory substance in the tumor-bearing
mice. They induced significant activation of the comple-
ment system, increased cytokine production, and led to
macrophage cell activation. All of these immune responses
elicited by the wsMWNT boosted the general activity of the
host immune system and modified the progression of the
H22 tumor [52].
CNTs have also shown the potential to boost the anti-
genicity of the carried proteins or peptides. We have inves-
tigated whether MWNTs conjugated to tumor lysate protein
will enhance the efficacy of an anti-tumor immunotherapy
that employs tumor cell vaccine (TCV) in a mouse model
bearing the H22 liver cancer. The study shows that MWNTs
conjugated to tumor lysate protein enhanced the specific
anti-tumor immune response and the cancer cure rate of a
TCV immunotherapy in mice. The results suggest that
MWNTs may play a role in the development of new anti-
tumor immunotherapies [53].
The interaction of CNTs with tumor-associated macro-
phage cells deserves attention. For example, to evaluate the
potential application of CNTs for brain tumor therapy, one
group studied the uptake and toxicity of MWNTs in the
GL261 murine intracranial glioma model. It is noteworthy
that most MWNTs were uptaken by tumor-associated
macrophages. This suggests that MWNTs may potentially
be used as a novel and non-toxic vehicle for targeting
macrophages in brain tumors [54].
6 Evaluation of the risks and benefits of CNT
based materials in cancer therapies
The potential applications for CNTs in cancer therapy have
attracted increasing interest in both research and clinics.
However, debate remains over the potential risks and bene-
fits of applying CNTs in antitumor therapies. One review
article has made a comparison between the characteristics,
advantages, drawbacks, benefits, and risks associated with
these novel biocompatible forms of carbon [55]. The inter-
esting property of being taken up by various kinds of cells
without inducing severe cytotoxicity has attracted intensive
research and development interests; however, only a few
6 XU HaiYan, et al. Sci China Chem November (2010) Vol.53 No.11
examples of small molecule delivery (antibacterial, antiviral,
and anticancer agents) using f-CNTs have been reported
previously. The authors believe that this could be attributed
to the fact that only a limited portion of the whole surface is
available, and thus the drug loading does not guarantee the
desired pharmacological effect. In addition, no application
for drug delivery has yet been approved or entered the mar-
ket, which favors an increased skepticism toward any
bio-application of these nanotubes. Nevertheless, many in-
teresting results may be expected in the very near future,
which shows that CNTs need to be investigated much more
deeply for their potential impact on nanoscience. Another
article analyzes the potential, through possible toxicological
implications, of CNTs in nanomedicine. The article pro-
poses that the lack of a centralized toxicity database limits
comparison between research results and that it is necessary
to understand deeply the postexposure regeneration, resis-
tance, and mechanisms of injury in cells due to CNTs be-
fore they can be used in medicine [56].
This work was supported by the National Basic Research Program of
China (2006CB933203 & 2010CB934002).
1 Liu Z, Chen K, Davis C, Sherlock S, Cao Q, Chen X, Dai H. Drug
delivery with carbon nanotubes for in vivo cancer treatment. Cancer
Res, 2008, 68(16): 6652–6660
2 Wu W, Li R, Bian X, Zhu Z, Ding D, Li X, Jia Z, Jiang X, Hu Y.
Covalently combining carbon nanotubes with anticancer agent: Prepa-
ration and antitumor activity. ACS Nano, 2009, 3(9): 2740–2750
3 Samorì C, Ali-Boucetta H, Sainz R, Guo C, Toma FM, Fabbro C, da
Ros T, Prato M, Kostarelos K, Bianco A. Enhanced anticancer activ-
ity of multi-walled carbon nanotube-methotrexate conjugates using
cleavable linkers. Chem Commun (Camb), 2010, 46(9): 1494–1496
4 Chaudhuri P, Soni S, Sengupta S. Single-walled carbon nanotube-
conjugated chemotherapy exhibits increased therapeutic index in
melanoma. Nanotechnology, 2010, 21(2): 025102
5 Ali-Boucetta H, Al-Jamal KT, McCarthy D, Prato M, Bianco A,
Kostarelos K. Multiwalled carbon nanotube-doxorubicin supramolecular
complexes for cancer therapeutics. Chem Commun (Camb), 2008, (4):
459–461
6 Liu Z, Fan AC, Rakhra K, Sherlock S, Goodwin A, Chen X, Yang Q,
Felsher DW, Dai H. Supramolecular stacking of doxorubicin on car-
bon nanotubes for in vivo cancer therapy. Angew Chem Int Ed, 2009,
48(41): 7668–7672
7 Lay CL, Liu HQ, Tan HR, Liu Y. Delivery of paclitaxel by physi-
cally loading onto poly(ethylene glycol) (PEG)-graft-carbon nano-
tubes for potent cancer therapeutics. Nanotechnology, 2010, 21(6):
065101
8 Murakami T, Sawada H, Tamura G, Yudasaka M, Iijima S, Tsuchida
K. Water-dispersed single-wall carbon nanohorns as drug carriers for
local cancer chemotherapy. Nanomedicine (Lond), 2008, 3(4):
453–463
9 Ajima K, Murakami T, Mizoguchi Y, Tsuchida K, Ichihashi T, Iijima
S, Yudasaka M. Enhancement of in vivo anticancer effects of cis-
platin by incorporation inside single-wall carbon nanohorns. ACS
Nano, 2008, 2(10): 2057–2064
10 Hampel S, Kunze D, Haase D, Krämer K, Rauschenbach M, Ritschel
M, Leonhardt A, Thomas J, Oswald S, Hoffmann V, Büchner B.
Carbon nanotubes filled with a chemotherapeutic agent: A nanocar-
rier mediates inhibition of tumor cell growth. Nanomedicine (Lond),
2008, 3(2): 175–182
11 Mahmood M, Karmakar A, Fejleh A, Mocan T, Iancu C, Mocan L,
Iancu DT, Xu Y, Dervishi E, Li Z, Biris AR, Agarwal R, Ali N,
Galanzha EI, Biris AS, Zharov VP. Synergistic enhancement of can-
cer therapy using a combination of carbon nanotubes and anti-tumor
drug. Nanomedicine (Lond), 2009, 4(8): 883–893
12 Weng X, Wang M, Ge J, Yu S, Liu B, Zhong J, Kong J. Carbon
nanotubes as a protein toxin transporter for selective HER2-positive
breast cancer cell destruction. Mol Biosyst, 2009, 5(10): 1224–1231
13 Bhirde AA, Patel V, Gavard J, Zhang G, Sousa AA, Masedunskas A,
Leapman RD, Weigert R, Gutkind JS, Rusling JF. Targeted killing of
cancer cells in vivo and in vitro with EGF-directed carbon nanotube-
based drug delivery. ACS Nano, 2009, 3(2): 307–316
14 Dhar S, Liu Z, Thomale J, Dai H, Lippard SJ. Targeted single-wall
carbon nanotube-mediated Pt(IV) prodrug delivery using folate as a
homing device. J Am Chem Soc, 2008, 130(34): 11467–11476
15 Yang F, Fu DL, Long J, Ni QX. Magnetic lymphatic targeting drug
delivery system using carbon nanotubes. Med Hypotheses, 2008,
70(4): 765–767
16 Zhang X, Meng L, Lu Q, Fei Z, Dyson PJ. Targeted delivery and
controlled release of doxorubicin to cancer cells using modified sin-
gle wall carbon nanotubes. Biomaterials, 2009, 30(30): 6041–6047
17 Li R, Wu R, Zhao L, Wu M, Yang L, Zou H. P-glycoprotein antibody
functionalized carbon nanotube overcomes the multidrug resistance
of human leukemia cells. ACS Nano, 2010, 4(3): 1399–1408
18 Moon HK, Lee SH, Choi HC. In vivo near-infrared mediated tumor
destruction by photothermal effect of carbon nanotubes. ACS Nano,
2009, 3(11): 3707–3713
19 Burlaka A, Lukin S, Prylutska S, Remeniak O, Prylutskyy Y, Shuba
M, Maksimenko S, Ritter U, Scharff P. Hyperthermic effect of
multi-walled carbon nanotubes stimulated with near infrared irradia-
tion for anticancer therapy: in vitro studies. Exp Oncol, 2010, 32(1):
48–50
20 Biris AS, Boldor D, Palmer J, Monroe WT, Mahmood M, Dervishi E,
Xu Y, Li Z, Galanzha EI, Zharov VP. Nanophotothermolysis of mul-
tiple scattered cancer cells with carbon nanotubes guided by time-
resolved infrared thermal imaging. J Biomed Opt, 2009, 14(2): 021007
21 Burke A, Ding X, Singh R, Kraft RA, Levi-Polyachenko N, Rylander
MN, Szot C, Buchanan C, Whitney J, Fisher J, Hatcher HC,
D'Agostino RJ, Kock ND, Ajayan PM, Carroll DL, Akman S, Torti
FM, Torti SV. Long-term survival following a single treatment of
kidney tumors with multiwalled carbon nanotubes and near-infrared
radiation. Proc Natl Acad Sci, 2009, 106(31): 12897–12902
22 Torti SV, Byrne F, Whelan O, Levi N, Ucer B, Schmid M, Torti FM,
Akman S, Liu J, Ajayan PM, Nalamasu O, Carroll DL. Thermal abla-
tion therapeutics based on CN(x) multi-walled nanotubes. Int J
Nanomedicine, 2007, 2(4): 707–714
23 Wang CH, Huang YJ, Chang CW, Hsu WM, Peng CA. In vitro pho-
tothermal destruction of neuroblastoma cells using carbon nanotubes
conjugated with GD2 monoclonal antibody. Nanotechnology, 2009,
20(31): 315101
24 Levi-Polyachenko NH, Merkel EJ, Jones BT, Carroll DL, Stewart JH
4th. Rapid photothermal intracellular drug delivery using multiwalled
carbon nanotubes. Mol Pharm, 2009, 6(4): 1092–1099
25 Klingeler R, Hampel S, Büchner B. Carbon nanotube based bio-
medical agents for heating, temperature sensoring and drug delivery.
Int J Hyperthermia, 2008, 24(6): 496–505
26 Marches R, Chakravarty P, Musselman IH, Bajaj P, Azad RN, Pan-
tano P, Draper RK, Vitetta ES. Specific thermal ablation of tumor
cells using single-walled carbon nanotubes targeted by covalently-
coupled monoclonal antibodies. Int J Cancer, 2009, 125(12):
2970–2977
27 Xiao Y, Gao X, Taratula O, Treado S, Urbas A, Holbrook RD,
Cavicchi RE. Avedisian Anti-HER2 IgY antibody-functionalized
single-walled carbon nanotubes for detection and selective destruc-
tion of breast cancer cells. BMC Cancer, 2009, 9: 351
28 Chakravarty P, Marches R, Zimmerman NS, Swafford AD, Bajaj P,
Musselman IH, Pantano P, Draper RK, Vitetta ES. Thermal ablation
of tumor cells with antibody-functionalized single-walled carbon
nanotubes. Proc Natl Acad Sci, 2008, 105(25): 8697–8702
29 Zhou F, Xing D, Ou Z, Wu B, Resasco DE, Chen WR. Cancer pho-
XU HaiYan, et al. Sci China Chem November (2010) Vol.53 No.11 7
tothermal therapy in the near-infrared region by using single-walled
carbon nanotubes. J Biomed Opt, 2009, 14(2): 021009
30 Kang B, Yu D, Dai Y, Chang S, Chen D, Ding Y. Cancer-cell target-
ing and photoacoustic therapy using carbon nanotubes as “bomb”
agents. Small, 2009, 5(11): 1292–1301
31 Kam NW, O'Connell M, Wisdom JA, Dai H. Carbon nanotubes as
multifunctional biological transporters and near-infrared agents for
selective cancer cell destruction. Proc Natl Acad Sci, 2005, 102(33):
11600–11605
32 Ghosh S, Dutta S, Gomes E, Carroll D, D'Agostino RJ, Olson J,
Guthold M, Gmeiner WH. Increased heating efficiency and selective
thermal ablation of malignant tissue with DNA-encased multiwalled
carbon nanotubes. ACS Nano, 2009, 3(9): 2667–2673
33 Gannon CJ, Cherukuri P, Yakobson BI, Cognet L, Kanzius JS, Kit-
trell C, Weisman RB, Pasquali M, Schmidt HK, Smalley RE, Curley
SA. Carbon nanotube-enhanced thermal destruction of cancer cells in
a noninvasive radiofrequency field. Cancer, 2007, 110(12):
2654–2665
34 Mashal A, Sitharaman B, Li X, Avti P, Sahakian A, Booske J, Hag-
ness S. Toward carbon-nanotube-based theranostic agents for micro-
wave detection and treatment of breast cancer: enhanced dielectric
and heating response of tissue-mimicking materials. IEEE Trans
Biomed Eng, 2010 Feb 18. [Epub ahead of print]
35 Cheung W, Pontoriero F, Taratula O, Chen AM, He H. DNA and
carbon nanotubes as medicine. Adv Drug Deliv Rev, 2010, 22
36 Bartholomeusz G, Cherukuri P, Kingston J, Cognet L, Lemos R,
Leeuw TK, Gumbiner-Russo L, Weisman RB, Powis G. In vivo
therapeutic silencing of hypoxia-inducible factor 1 alpha (HIF-1alpha)
using single-walled carbon nanotubes noncovalently coated with
siRNA. Nano Res, 2009, 2(4): 279–291
37 Podesta JE, Al-Jamal KT, Herrero MA, Tian B, Ali-Boucetta H,
Hegde V, Bianco A, Prato M, Kostarelos K. Antitumor activity and
prolonged survival by carbon-nanotube-mediated therapeutic siRNA
silencing in a human lung xenograft model. Small, 2009, 5(10):
1176–1185
38 Wang X, Ren J, Qu X. Targeted RNA interference of cyclin A2 me-
diated by functionalized single-walled carbon nanotubes induces pro-
liferation arrest and apoptosis in chronic myelogenous leukemia
K562 cells. ChemMedChem, 2008, 3(6): 940–945
39 Pan B, Cui D, Xu P, Ozkan C, Feng G, Ozkan M, Huang T, Chu B,
Li Q, He R, Hu G. Synthesis and characterization of polyamidoamine
dendrimer-coated multi-walled carbon nanotubes and their applica-
tion in gene delivery systems. Nanotechnology, 2009, 20(12): 125101
40 Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes
in drug delivery. Curr Opin Chem Biol, 2005, 9(6): 674–679
41 Berd D, Sato T, Maguire HC, Kairys JJ, Mastrangelo MJ. Immuno-
pharmacologic analysis of an autologous, hapten-modified human
melanoma vaccine. J Clin Oncol, 2004, 22(3): 403–415
42 Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccina-
tion with tumor lysate-pulsed dendritic cells elicits antigen-specific,
cytotoxic T-cells in patients with malignant glioma. Cancer Res,
2004, 64(14): 4973–4979
43 Cai D, Mataraza JM, Qin ZH, Huang Z, Huang J, Chiles TC,
Carnahan D, Kempa K, Ren Z. Highly efficient molecular delivery
into mammalian cells using carbon nanotube spearing. Nat Methods,
2005, 2: 449–454
44 Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber
JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of
283 consecutive patients with metastatic melanoma or renal cell can-
cer using high-dose bolus interleukin 2. Jama, 1994, 271(12):
907–913
45 Rosenberg SA, Yang JC, White DE, Steinberg SM. Durability of
complete responses in patients with metastatic cancer treated with
high-dose interleukin-2: Identification of the antigens mediating re-
sponse. Ann Surg, 1998, 228(3): 307–319
46 Chatterjee M, Draghici S, Tainsky MA. Immunotheranostics: Break-
ing tolerance in immunotherapy using tumor autoantigens identified
on protein microarrays. Curr Opin Drug Discov Devel, 2006, 9(3):
380–385
47 Muller AJ, Scherle PA. Targeting the mechanisms of tumoral
immune tolerance with small-molecule inhibitors. Nat Rev Cancer,
2006, 6(1): 613–625
48 Copier J, Dalgleish A. Overview of tumor cell-based vaccines. Int
Rev Immunol, 2006, 25(5&6): 297–319
49 Banchereau J, Schuler-Thurner B, Palucka AK, Schuler G. Dendritic
Cells as vectors for therapy. Cell, 2001, 106(3): 271–274
50 Emens LA. Roadmap to a Better Therapeutic tumor vaccine. Int Rev
Immunol, 2006, 25(5/6): 415–443
51 Schirrmacher V. Clinical trials of antitumor vaccination with an
autologous tumor cell vaccine modified by virus infection: Improve-
ment of patient survival based on improved antitumor immune mem-
ory. Cancer Immunol, Immunother, 2005, 54(6): 587–598
52 Meng J, Yang M, Jia F, Kong H, Zhang WQ, Wang CY, Xing JM,
Xie SS, Xu HY. Subcutaneous injection of water-soluble multi-
walled carbon nanotubes in tumor-bearing mice boosts the host im-
mune activity. Nanotechnology, 2010, 21(14): 145104
53 Meng J, Meng J, Duan J, Kong H, Li L, Wang C, Xie S, Chen S, Gu
N, Xu H, Yang XD. Carbon nanotubes conjugated to tumor lysate
protein enhance the efficacy of an antitumor immunotherapy. Small,
2008, 4(9): 1364–1370
54 VanHandel M, Alizadeh D, Zhang L, Kateb B, Bronikowski M,
Manohara H, Badie B. Selective uptake of multi-walled carbon
nanotubes by tumor macrophages in a murine glioma model. J
Neuroimmunol, 2009, 208(1-2): 3–9
55 Pastorin G. Crucial functionalizations of carbon nanotubes for im-
proved drug delivery: A valuable option? Pharm Res, 2009, 26(4):
746–769
56 Firme CP 3rd, Bandaru PR. Toxicity issues in the application of car-
bon nanotubes to biological systems. Nanomedicine, 2010, 6(2):
245–256
... 132 Results showed that CNTs are excellent candidates for immunomodulation, enhancing antitumor immune activity in murine models. 133 Oxidized and subcutaneously administered CNTs promote activation of the complement system, activation of inflammatory cytokines and macrophages, resulting in inhibition of tumor growth. 134 In this regard, there is growing interest in the use of CNTs for immunotherapeutic applications. ...
Article
Full-text available
Since December 2019, the world has faced an unprecedented pandemic crisis due to a new coronavirus disease, coronavirus disease-2019 (COVID-19), which has instigated intensive studies on prevention and treatment possibilities. Here, we investigate the relationships between the immune activation induced by three coronaviruses associated with recent outbreaks, with special attention to SARS-CoV-2, the causative agent of COVID-19, and the immune activation induced by carbon nanotubes (CNTs) to understand the points of convergence in immune induction and modulation. Evidence suggests that CNTs are among the most promising materials for use as immunotherapeutic agents. Therefore, this investigation explores new possibilities of effective immunotherapies for COVID-19. This study aimed to raise interest and knowledge about the use of CNTs as immunotherapeutic agents in coronavirus treatment. Thus, we summarize the most important immunological aspects of various coronavirus infections and describe key advances and challenges in using CNTs as immunotherapeutic agents against viral infections and the activation of the immune response induced by CNTs, which can shed light on the immunotherapeutic possibilities of CNTs.
... In recent years, targeted drug delivery systems based on nano-particles have been reported to provide promising directions to overcome the above-mentioned challenges. [1] Nano particles, which exhibit unusual physical and chemical properties, have gained more interest in different disciplines and have been applied to a wide variety of fields, including biological and biomedical engineering. Nanoparticles have a great potential as a carrier of various active drugs at the target sites. ...
Article
Full-text available
The green synthesized zinc oxide nanoparticle (ZnO) was filled with a commercially available anti‐cancer drug, Doxorubicin (DoX). The loading of the drug into ZnO was validated using UV‐Visible and Raman spectroscopy. X‐ray diffraction data showed the wurtzite structure of the nanoparticles of ZnO. Scanning electron microphotography images detected the encapsulation of DoX drugs into ZnO nanoparticles. Fourier transformer infrared spectrum of DoX loaded ZnO showed a single band at 478 cm⁻¹ attributed to the ZnO vibrational band and the carbonyl group peak at 1578 cm⁻¹, suggesting the loading of DoX into ZnO. In addition, in vitro cytotoxicity of DoX, ZnO and ZnO‐DoX against HeLa cells was studied using the MTT colorimetric cell viability assay, which showed 99.4 % inhibition of ZnO‐DoX cells at a concentration of 100 μg/mL. The findings thus provide compelling evidence for green biosynthesized ZnO, which could serve as a promising nano‐drug carrier for the targeted drug delivery system.
... Advanced nanotechnology has been widely utilized in the treatment of cancer [55][56][57]. Due to their unique physiochemical properties, CNTs have been exploited as multipurpose innovative carriers for cancer therapy [58,59]. ...
Chapter
There has been a predominant interest in using nanotechnology for drug delivery applications, especially for cite-specific targeting. Recently, interest in the potential of carbon nanotubes (CNT) to deliver bio-molecules for a range of biomedical applications, particularly drug delivery into living systems for cancer diagnostics and therapy has massively grown. Pure CNTs have inherent limitations, such as poor solubility, which limit their use in biomedical applications. A myriad of approaches to enhance their solubility have been investigated. An attractive approach has involved the functionalization of CNTs (f-CNT) in order to improve their solubility and biocompatibility in aqueous solutions. This chapter provides an introductory overview of carbon nanotubes and their use as drug delivery systems for cancer treatment.
Article
Brain cancers, especially glioblastoma multiforme (GBM), are associated with poor prognosis due to the limited efficacy of current therapies. Nanomedicine has emerged as a versatile technology to treat various diseases, including cancers, and has played an indispensable role in combatting the COVID‐19 pandemic as evidenced by the role that lipid nanocarrier‐based vaccines have played. The tunability of nanocarrier physicochemical properties – including size, shape, surface chemistry, and drug release kinetics – has resulted in the development of a wide range of nanocarriers for brain cancer treatment. These nanocarriers can improve the pharmacokinetics of drugs, increase blood‐brain barrier (BBB) transfer efficiency, and specifically target brain cancer cells. These unique features would potentially allow for more efficient treatment of brain cancer with less side effects and better therapeutic outcomes. This review provides an overview of brain cancers, current therapeutic options, and challenges to efficient brain cancer treatment. The latest advances in nanomedicine strategies are investigated with an emphasis on targeted and stimulus‐responsive nanocarriers and their potential for clinical translation. This article is protected by copyright. All rights reserved
Article
Carbon nanotubes (CNTs) have been largely investigated in various biomedical fields on the basis of their excellent physical properties and drug delivery performance. However, application capacities of CNTs in blood-contacting medical devices are given due attention though there have been increasingly accumulated experimental data showing promising potentials. Herein, we collected and showed research evidence that strong interactions of CNTs to plasm proteins are attractive and valuable features holding great application potentials for medical devices and implants used in blood-contacting environments, while blood compatibility has been a big challenge faced by this kind of devices. This review introduces the strong and nonspecific plasm protein adsorptions of CNTs due to their high purity of carbon composition and nanostructures, followed by discussions on the implication of these interactions to blood coagulation and complement activation, aiming to sort out and provide insights into the application potentials of CNTs in blood-contacting medical devices and implants in the context of anti-thrombosis and blood purification.
Chapter
Radio frequency reactive magnetron sputtering was used to produce silver oxide thin films for antimicrobial applications. The morphology and bandgap of the films were tuned using oxygen flow rate and forward deposition power to obtain monolithic, transparent, visible-light activated photocatalyst. XRD and RDF indicated the deposition of antimicrobial phases of silver oxide (Ag4O4 and Ag2O) nanocrystalline particles (2.45–31.30 nm) in a mixture of amorphous and crystalline background typical of films deposited at room temperature as confirmed by SEM images. Contact angle and surface energy analysis indicated the films are mildly hydrophilic and the Lifshitz-van der Walls forces as well as polarization of the surfaces providing interactive forces on the films. Optical characterization reveals absorption in the visible spectrum leading to photocatalysis as well as transmission of up to 80% of visible light and a bandgap range of 2.35–3.15 eV for the films. Atomic absorption spectroscopy confirmed 24 hours sustained silver ion release in both water and saline. XPS analysis confirmed photocatalysis on the surfaces capturing superoxide and hydroxyl groups on the films. Antimicrobial tests confirmed 100% killing of both Gram-positive and Gram-negative bacteria in less than 25 minutes. The findings open new doors for the development of non-composite visible-light activated photocatalyst applications.
Article
Carbon nanotubes (CNTs) are graphene sheets of nanometer size with unique strength and physio-chemical properties. These nano-structures of single and multi-layer graphene sheets have potential to conjugate with various functional groups. Recent advances in life sciences and nanotechnology have revealed that salient features of CNTs enable them to play promising role in diverse areas of health like drug delivery, gene therapy, cancer treatment, artificial implants, tissue regeneration, and neurodegenerative diseases. This review focuses on the comprehensive understanding of applications of CNTs in medicinal therapy. The striking applications associated with CNTs cannot be denied; however, there are certain peculiar potential drawbacks associated with CNTs in form of toxic effects like asthma, lung disorders, renal, and hepatic malfunctioning as well as neural damage. Due to their prime role in medical science, further research should be done on these evolutionary nanoparticles to have more efficacious and safe CNTs for welfare of humanity.
Chapter
This chapter gives an overview of applications of photothermal spectroscopy to chemical analysis when the sample has been mostly prepared in homogeneous form. There are two main applications of photothermal spectroscopy to chemical analysis of low absorption analytes. The first is analysis of stationary samples. In this application, analysis samples are introduced into the photothermal apparatus, and the photothermal signal is subsequently measured. The second application for measurement of optical absorption is dynamic samples. Dynamic samples exhibit a change in analyte absorbance as a function of time. An important trend in the development of analytical chemistry is the miniaturization of instruments and chemical analysis techniques in combination with the integration of all stages of analysis (sampling, sample preparation, separation, preconcentration, analytical reactions, and analytical signal measurement).
Chapter
Full-text available
Photothermal cancer therapy, also called photothermal therapy (PTT), uses lightinduced heat to treat cancer cells. PTT has attracted attention owing to its advantages over traditional cancer therapies such as chemotherapy, radiation therapy (RT), and surgery. Thermal therapy was discovered during the nineteenth century when cancer patients were administered living bacteria to cause inflammation and subsequent fever. The use of circulating heated water to treat uterine cervical cancer was also reported during that era. Owing to the rudimentary technology of that age, neither clinical applications nor further studies of thermal therapy were explored. Interest in PTT was revived in the 1980s, and owing to the fast development of optical and nanomaterial technology during recent decades, thermal therapy, particularly PTT has become a fast‐growing field of study [1–5].
Article
The thermal lens technique is proposed for determination of total haemoglobin in the form of reversible hemichrome. The conditions were optimized (concentration of sodium dodecyl sulfate, 2 mM) to attain the maximum sensitivity with the use of polyethylene glycols as signal enhancers. For polyethylene glycols with molecular weights 1500|–|35000 Da in a concentration range of 5 to 15% w/w (5 to 25 mM), the influence on thermal lens signal enhancement was estimated. It is shown that the use of 5% w/w polyethylene glycol 2000 provides the maximum increase in the thermal-lens enhancement factor (by 40%) in comparison with unmodified aqueous solutions. The detection limit of iron(II) tris(1,10-phenanthrolinate) as a model system is 60 nM. Under these conditions, the thermal-lens detection limit of hemichrome is 10 nM, which shows a 15-fold enhancement compared to spectrophotometry. Modification of the medium with polyethylene glycols decreases the limit of detection of hemichrome determination by 15% in comparison with unmodified aqueous solutions due to better reproducibility for the range of concentrations from 0.02 to 0.9 µM.
Article
Full-text available
The experimental results reported in this paper suggest that single-walled carbon nanotubes (SWCNTs) have the potential to enhance dielectric contrast between malignant and normal tissue for microwave detection of breast cancer and facilitate selective heating of malignant tissue for microwave hyperthermia treatment of breast cancer. In this study, we constructed tissue-mimicking materials with varying concentrations of SWCNTs and characterized their dielectric properties and heating response. At SWCNT concentrations of less than 0.5% by weight, we observed significant increases in the relative permittivity and effective conductivity. In microwave heating experiments, we observed significantly greater temperature increases in mixtures containing SWCNTs. These temperature increases scaled linearly with the effective conductivity of the mixtures. This work is a first step towards the development of functionalized, tumor-targeting SWCNTs as theranostic (integrated therapeutic and diagnostic) agents for microwave breast cancer detection and treatment.
Article
Full-text available
The identification of disease-related genes and their complete nucleotide sequence through the human genome project provides us with a remarkable opportunity to combat a large number of diseases with designed genes as medicine. However, gene therapy relies on the efficient and nontoxic transport of therapeutic genetic medicine through the cell membranes, and this process is very inefficient. Carbon nanotubes, due to their large surface areas, unique surface properties, and needle-like shape, can deliver a large amount of therapeutic agents, including DNA and siRNAs, to the target disease sites. In addition, due to their unparalleled optical and electrical properties, carbon nanotubes can deliver DNA/siRNA not only into cells, which include difficult transfecting primary-immune cells and bacteria, they can also lead to controlled release of DNA/siRNA for targeted gene therapy. Furthermore, due to their wire shaped structure with a diameter matching with that of DNA/siRNA and their remarkable flexibility, carbon nanotubes can impact on the conformational structure and the transient conformational change of DNA/RNA, which can further enhance the therapeutic effects of DNA/siRNA. Synergistic combination of the multiple capabilities of carbon nanotubes to deliver DNA/siRNAs will lead to the development of powerful multifunctional nanomedicine to treat cancer or other difficult diseases. In this review, we summarized the current studies in using CNT as unique vehicles in the field of gene therapy.
Article
Nature Reviews. Cancer 6, 613–625 (2006) There is an error in Figure 1 of this article on page 616. The mature immunogenic dendritic cell at the top of the figure should not be expressing IDO. The correct version of the figure is shown below.
Article
Objective. —To determine the efficacy of treatment using high-dose bolus Interleukin 2 (IL-2) in patients with metastatic melanoma or renal cell cancer.Design and Setting. —Consecutive series of all patients treated with high-dose IL-2 in the Surgery Branch of the National Cancer Institute from September 1985 through December 1992.Patients. —Two hundred eighty-three patients with metastatic melanoma or metastatic renal cell cancer who had failed standard treatment for their cancers.Interventions. —Patients received IL-2 at a dose of 720 000 lU/kg intravenously every 8 hours for a maximum of 15 doses per cycle. Two cycles constituted a treatment course, and patients with stable or responding disease received additional treatment courses. A total of 447 courses of treatment were administered.Main Outcome Measures. —Regression of measurable tumor, durability of response to treatment, and survival.Results. —Nine patients (7%) with metastatic melanoma achieved complete regression of all disease and 14 patients (10%) had partial regression. Ten patients (7%) with metastatic renal cell cancer experienced complete regression and 20 patients (13%) had partial regression. Of the 19 patients with complete regression, 15 have remained in complete remission from 7 to 91 months after treatment. Three treatment-related deaths (1.1%) occurred early in this series, but as experience with the administration of this IL-2 regimen increased, no treatment-related deaths occurred in 214 patients treated during the last 5 years of the study.Conclusion. —Biologic therapy with IL-2 can cause significant antitumor effects in patients with advanced metastatic melanoma or renal cell cancer. Because IL-2 does not have a direct effect on cancer cells but rather mediates its antitumor activity by altering host immune reactions, these data represent the best available evidence that immunologic therapy for cancer can be effective in selected patients.(JAMA. 1994;271:907-913)
Article
Carbon nanotubes are unique materials that absorb infrared (IR) radiation, especially between 700 and 1100 nm, where body tissues are most transparent. Absorbed IR promotes molecular oscillation leading to efficient heating of the surrounding environment. A method to enhance drug localization for peritoneal malignancies is perfusion of warm (40−42 °C) chemotherapeutic agents in the abdomen. However, all tissues in the peritoneal cavity are subjected to enhanced drug delivery due to increased cell membrane permeability at hyperthermic temperatures. Here we show that rapid heating (within ten seconds) of colorectal cancer cells to 42 °C, using infrared stimulation of nanotubes as a heat source, in the presence of the drugs oxaliplatin or mitomycin C, is as effective as two hours of radiative heating at 42 °C for the treatment of peritoneal dissemination of colorectal cancer. We demonstrate increased cell membrane permeability due to hyperthermia from multiwalled carbon nanotubes in close proximity to cell membranes and that the amount of drug internalized by colorectal cancer cells heated quickly using carbon nanotubes equals levels achieved during routine application of hyperthermia at 42 °C. This approach has the potential to be used as a rapid bench to bedside clinical therapeutic agent with significant impact for localizing chemotherapy agents during the surgical management of peritoneal dissemination of colorectal cancer.Keywords: Hyperthermia; multiwalled carbon nanotubes; colorectal cancer
Article
Single-walled carbon nanotubes (SWNTs) have remarkable physicochemical properties that may have several medical applications. The authors have discovered a novel property of SWNTs—heat release in a radiofrequency (RF) field—that they hypothesized may be used to produce thermal cytotoxicity in malignant cells. Functionalized, water-soluble SWNTs were exposed to a noninvasive, 13.56-megahertz RF field, and heating characteristics were measured with infrared thermography. Three human cancer cell lines were incubated with various concentrations of SWNTs and then treated in the RF field. Cytotoxicity was measured by fluorescence-activated cell sorting. Hepatic VX2 tumors in rabbits were injected with SWNTs or with control solutions and were treated in the RF field. Tumors were harvested 48 hours later to assess viability. The RF field induced efficient heating of aqueous suspensions of SWNTs. This phenomenon was used to produce a noninvasive, selective, and SWNT concentration-dependent thermal destruction in vitro of human cancer cells that contained internalized SWNTs. Direct intratumoral injection of SWNTs in vivo followed by immediate RF field treatment was tolerated well by rabbits bearing hepatic VX2 tumors. At 48 hours, all SWNT-treated tumors demonstrated complete necrosis, whereas control tumors that were treated with RF without SWNTs remained completely viable. Tumors that were injected with SWNTs but were not treated with RF also were viable. The current results suggested that SWNTs targeted to cancer cells may allow noninvasive RF field treatments to produce lethal thermal injury to the malignant cells. Now, the authors are developing SWNTs coupled with cancer cell-targeting agents to enhance SWNT uptake by cancer cells while limiting uptake by normal cells. Cancer 2007.
Article
Most low-molecular-weight platinum anticancer drugs have short blood circulation times that are reflected in their reduced tumor uptake and intracellular DNA binding. A platinum(IV) complex of the formula c, c, t-[Pt(NH 3) 2Cl 2(O 2CCH 2CH 2CO 2H)(O 2CCH 2CH 2CONH-PEG-FA)] ( 1), containing a folate derivative (FA) at an axial position, was prepared and characterized. Folic acid offers a means of targeting human cells that highly overexpress the folate receptor (FR). Compound 1 was attached to the surface of an amine-functionalized single-walled carbon nanotube (SWNT-PL-PEG-NH 2) through multiple amide linkages to use the SWNTs as a "longboat delivery system" for the platinum warhead, carrying it to the tumor cell and releasing cisplatin upon intracellular reduction of Pt(IV) to Pt(II). The ability of SWNT tethered 1 to destroy selectively FR(+) vs FR(-) cells demonstrated its ability to target tumor cells that overexpress the FR on their surface. That the SWNTs deliver the folate-bearing Pt(IV) cargos into FR(+) cancer cells by endocytosis was demonstrated by the localization of fluorophore-labeled SWNTs using fluorescence microscopy. Once inside the cell, cisplatin, formed upon reductive release from the longboat oars, enters the nucleus and reacts with its target nuclear DNA, as determined by platinum atomic absorption spectroscopy of cell extracts. Formation of the major cisplatin 1,2-intrastrand d(GpG) cross-links on the nuclear DNA was demonstrated by use of a monoclonal antibody specific for this adduct. The SWNT-tethered compound 1 is the first construct in which both the targeting and delivery moieties have been incorporated into the same molecule; it is also the first demonstration that intracellular reduction of a Pt(IV) prodrug leads to the cis-{Pt((NH 3) 2} 1,2-intrastrand d(GpG) cross-link in nuclear DNA.