Anda di halaman 1dari 12

Lasers Med Sci (2008) 23:217–228

DOI 10.1007/s10103-007-0470-x

REVIEW ARTICLE

Plasmonic photothermal therapy (PPTT)


using gold nanoparticles
Xiaohua Huang & Prashant K. Jain & Ivan H. El-Sayed &
Mostafa A. El-Sayed

Received: 26 April 2007 / Accepted: 10 May 2007 / Published online: 3 August 2007
# Springer-Verlag London Limited 2007

Abstract The use of lasers, over the past few decades, has viz. plasmonic photothermal therapy (PPTT). The synthetic
emerged to be highly promising for cancer therapy tunability of the optothermal properties and the bio-
modalities, most commonly the photothermal therapy targeting abilities of the plasmonic gold nanostructures
method, which employs light absorbing dyes for achieving make the PPTT method furthermore promising. In this
the photothermal damage of tumors, and the photodynamic review, we discuss the development of the PPTT method
therapy, which employs chemical photosensitizers that with special emphasis on the recent in vitro and in vivo
generate singlet oxygen that is capable of tumor destruc- success using gold nanospheres coupled with visible lasers
tion. However, recent advances in the field of nanoscience and gold nanorods and silica–gold nanoshells coupled with
have seen the emergence of noble metal nanostructures near-infrared lasers.
with unique photophysical properties, well suited for
applications in cancer phototherapy. Noble metal nano- Keywords Surface plasmon resonance (SPR) .
particles, on account of the phenomenon of surface Plasmonic photothermal therapy (PPTT) . Cancer .
plasmon resonance, possess strongly enhanced visible and Gold nanospheres . Gold nanorods . Gold nanoshells .
near-infrared light absorption, several orders of magnitude Immunotargeting
more intense compared to conventional laser phototherapy
agents. The use of plasmonic nanoparticles as highly
enhanced photoabsorbing agents has thus introduced a
much more selective and efficient cancer therapy strategy, Introduction

The use of heat has become one of the major methods


for tumor therapy since its ancient usage in 1700 BC
X. Huang : P. K. Jain : M. A. El-Sayed (*) when a glowing tip of a firedrill was used for breast cancer
Laser Dynamics Laboratory, therapy [1]. Later heating sources ranging from radio-
School of Chemistry and Biochemistry,
frequency [2–5] to microwaves [6–9] as well as ultrasound
Georgia Institute of Technology,
Atlanta, GA 30332-0400, USA waves [10–12] were introduced to induce moderate heating
e-mail: melsayed@gatech.edu in a specific target region, which is termed as hyperthermia.
Hyperthermia is commonly defined as heating tissue to a
I. H. El-Sayed
temperature in the range 41–47°C for tens of minutes [13].
Department of Otolaryngology—Head and Neck Surgery,
Comprehensive Cancer Center, Tumors are selectively destroyed in this temperature range
University of California at San Francisco, because of their reduced heat tolerance compared to normal
San Francisco, CA 94143, USA tissue, which is due to their poor blood supply. Hyper-
thermia causes irreversible cell damage by loosening cell
M. A. El-Sayed
University of California, membranes and denaturing proteins. But the applications of
Berkeley, CA 94720, USA the heating sources conventionally employed for hyperther-
218 Lasers Med Sci (2008) 23:217–228

mia are limited because of their damage to surrounding [54], have been introduced. A major drawback of PDT is
healthy tissues. that the photosensitizing drug stays in the body for a long
A revolution in cancer therapy has taken place by the time, rendering the patient to be highly sensitive to light.
emerging use of laser light to achieve controlled and An alternative to PDT is the photothermal therapy
confined thermal damage in the tumor tissue. Laser, the (PTT) in which photothermal agents are employed to
acronym for light amplification by the stimulated emission achieve the selective heating of the local environment
of radiation [14], is an optical source that emits photons in a [55–61]. When the PTT agents absorbs light, electrons
coherent and narrow beam. It was proposed in 1959 [14] make transitions from the ground state to the excited state.
and first demonstrated in 1960 [15]. Laser usage in surgery The electronic excitation energy subsequently relaxes
was first reported by ophthalmologists in 1963 [16] and through nonradiative decay channels. This results in the
then reported for tumor eradication in 1965 [17] followed increase in the kinetic energy leading to the overheating of
by wide interest in late 1960s [18–20]. The laser light, the local environment around the light absorbing species.
usually neodymium–yttrium aluminum garnet (Nd–YAG, The heat produced can be employed for local cell or tissue
1.06 um) and CO2 laser (10.6 um) [21–25] can either be destruction [62–65]. The photoabsorbing agents can be
transmitted from optical fiber tip to exposed tumors in the natural chromophores in the tissue [66–71] or externally
air or delivered into a confined space by inserting the bare added dye molecules such as indocyanine green [72, 73],
end of the fiber into the center of the target tumor, which is naphthalocyanines [74], and porphyrins coordinated with
often called interstitial laser hyperthermia [26–30]. Laser transition metals [75]. Natural chromophores, however,
light has the characteristics of monochromaticity, coher- suffer from very low absorption. The choice of the
ence, and collimation [31–33]. These properties provide a exogenous photothermal agents is made on the basis of
narrow beam of high intensity, which transmits deep down their strong absorption cross sections and highly efficient
into the target tissue with minimal power loss and great light-to-heat conversion. This greatly reduces the amount of
precision. The biggest disadvantage of laser therapy is its laser energy required to achieve the local damage of the
nonselectivity. Both normal and tumor cells in the path of diseased cells, rendering the therapy method less invasive.
the laser light are damaged. The requirement of the high But the problem with dye molecules is their photobleaching
power density is another problem. High power laser output under laser irradiation.
up to tens to hundreds of watts has to be used to efficiently In recent years, the tremendous development of nano-
induce the tumor oblation [23]. technology has provided a variety of nanostructures with
Another type of tumor therapy method is the photody- unique optical properties that are useful in biology and
namic therapy (PDT), also known as photochemotherapy biomedicinal applications [76–83]. From the point of the
[34–41], which involves cell destruction caused by means view of cancer therapeutics, noble metal nanoparticles
of toxic singlet oxygen and/or other free radicals that are become very useful as agents for PTT on account of their
produced from a sequence of photochemical and photobi- enhanced absorption cross sections, which are four to five
ological processes. These processes are initiated by the orders of magnitude larger than those offered by conven-
reaction of a photosensitizer with tissue oxygen upon tional photoabsorbing dyes. This strong absorption ensures
exposure to a specific wavelength of light in the visible or effective laser therapy at relatively lower energies rendering
near-infrared (NIR) region. The earliest sensitizer used was the therapy method minimally invasive. Additionally, metal
acridine, which was reported in 1900 to kill paramecia [42] nanostructures have higher photostability, and they do not
and followed by eosin for skin cancer treatment in 1903 suffer from photobleaching. Currently, gold nanospheres
[43]. Although many chemicals have been later reported for [84–91], gold nanorods [92–95], gold nanoshells [96–99],
photochemical therapy, porphyrin-based sensitizers [44–49] gold nanocages [100, 101], and carbon nanotubes [102] are
lead the role in clinical applications because of their the chief nanostructures that have been demonstrated in
preferential retention in cancer tissues and due to the high photothermal therapeutics due to their strongly enhanced
quantum yields of singlet oxygen produced. The Photofrin® absorption in the visible and NIR regions on account of
[50], which is a purified hematoporphyrin derivative, has their surface plasmon resonance (SPR) oscillations. Of
been approved for clinic trials by the US Food and Drug these structures, the first three nanostructures (see Fig. 1)
Administration. Porphyrin-based therapy can only be used are especially promising because of their ease of prepara-
for tumors on or just under the skin or on the lining of tion, ready multi-functionalization, and tunable optical
internal organs or cavities because it absorbs light shorter properties. In the present review, we discuss the photo-
than 640 nm in wavelength. For deep-seated tumors, thermal properties of these plasmonic nanostructures and
second generation sensitizers, which have absorbance in their application in selective PTT. We propose the name
the NIR region, such as core-modified porphyrins [51], plasmonic photothermal therapy (PPTT) for this treatment
chlorins [52] phthalocyanine [53], and naphthalocyanine to distinguish it from PTT and PDT.
Lasers Med Sci (2008) 23:217–228 219

Fig. 1 Plasmonic gold nanostructures commonly used for PPTT. a reproduced with permission from [115]); c Nanoshells (TEM image
Nanospheres (transmission electron microscopy [TEM] image repro- reproduced with permission from [97])
duced with permission from [110]; b nanorods (TEM image

Photothermal properties of plasmonic gold 520 nm due to the transverse electronic oscillation [109,
nanoparticles 111, 112, 114–118]. Unlike spherical nanoparticles, the
absorption spectrum of the gold nanorods is very sensitive
In 1857, Faraday [103] made colloidal gold for the first to the aspect ratio (length/width). With an increase in the
time by reducing gold chloride with phosphors and nanorod aspect ratio, the SPR absorption wavelength
recognized that the reddish color was due to the small size maximum of the longitudinal band significantly redshifts
of the colloidal gold particles. In 1951, Turkevich et al. (Fig. 2b). Similarly, when the solid gold nanospheres are
[104] simplified the method by using sodium citrate as changed to gold shell structures, the absorption maximum
reducing agents. Since then, the interaction between light also greatly redshifts. In 1998, Halas and coworkers at Rice
and gold nanoparticles has been widely studied [105–112]. University [119] developed the gold nanoshell structure,
Gold nanoparticles absorb light strongly in the visible which is composed of a silica core (100–200 nm in diameter)
region due to the coherent oscillations of the metal surrounded by a thin layer of gold shells (5–20 nm). The
conduction band electrons in strong resonance with visible nanoshells absorb and scatter strongly in the NIR region
frequencies of light. This phenomenon is known as the SPR [120]. The optical resonance of the nanoshells can be tuned
[105–113]. The SPR frequency is dependent on the type of by adjusting the ratio of the thickness of the gold shell to
the metal, the size and shape of the metal nanoparticles, as the diameter of the silica core (Fig. 2c). It has been shown
well as the dielectric constant of the surrounding medium, that the smaller this ratio, the more redshifted is the SPR
thus imparting a unique optical tunability to the nanostruc- wavelength [97].
tures. When the size increases, the surface plasmon ab- The photothermal properties of gold nanoparticles have
sorption maximum slightly redshifts (Fig. 2a). When the been systematically studied using femtosecond transient
nanoparticles form assemblies or aggregates, the surface absorption spectroscopy by Link and El-Sayed [111], who
plasmon absorption maximum redshifts to the NIR region. have shown that the photoexcitation of metal nanostruc-
Interestingly, when the shape of the gold nanoparticles is tures results in the formation of a heated electron gas that
changed from sphere to rod, the SPR spectrum splits into subsequently cools rapidly within ∼1 ps by exchanging
two bands: a stronger long-wavelength band in the NIR energy with the nanoparticle lattice. This is followed by
region due to the longitudinal oscillation of electrons and a phonon–phonon interactions where the nanoparticle lattice
weaker short-wavelength band in the visible region around cools rapidly by exchanging energy with the surrounding
220 Lasers Med Sci (2008) 23:217–228

mine 6G dye molecules [122]. Hot electron temperatures of


several thousand kelvins are easily reached in the nano-
particles even with laser excitation powers as low as 100 nJ
and the lattice temperature on the order of a few tens of
degrees can be achieved [111]. This highly efficient pro-
duction of heat energy from the absorbed light energy by
gold nanoparticles make them greatly promising in the
PPTT of cancers and other diseases. Further, in the case of
gold nanorods and gold nanoshells, this strong absorption can
be tuned to the NIR region (Fig. 2b,c), a region where light
penetration is optimal due to minimal absorption from tissue
chromophores and water [123]. This makes NIR-resonant
gold nanostructures very useful for clinical therapy applica-
tions involving tumors located deep within bodily tissue.
In addition to the local heating of the surrounding
environment, which leads to irreversible cell destruction
through protein denaturation and coagulation as well as cell
membrane destruction, bubble formation around gold
nanoparticles is also involved in the case of short pulse
laser irradiation, which imposes mechanical stress leading
to cell damage. Irradiation with short laser pulses has been
shown to lead to the rapid heating of the particles and va-
porization of a thin layer of fluid surrounding each particle,
producing a microscopic version of underwater explosion
and cavitation bubble formation [84–86, 88, 124, 125].
Zharov et al. [88] also found that nanoclusters formed by the
assembly of gold nanoparticles on human breast cancer cells
significantly enhance the bubble formation causing more
efficient cancer cell killing.
Very recently Khlebtsov et al. theoretically simulated the
photothermal conversion efficiency of the different nano-
structures including gold nanospheres, gold nanorods, gold
nanoshells, linear chains, 2D arrays, and 3D clusters [91]
by calculating their SPR absorption spectra. It was found
that gold spheres with diameters of about 30–40 nm are
most preferable, as their normalized absorption is maximal
in the visible spectrum region. The nanorods with length
between 15 and 70 nm were predicted to be most efficient.
Fig. 2 Size, shape, and composition dependence of the surface plasmon
absorption spectrum of plasmonic gold nanostructures. a Nanospheres of
Of course, it would also be required that the longitudinal
different sizes (reproduced with permission from [110]); b nanorods of absorption maximum be matched to the wavelength of the
different aspect ratios (reproduced with permission from [118]); c nano- NIR laser to get optimal photothermal efficiency. Gold
shells of different shell thicknesses (reproduced with permission from nanoshells with external diameters of about 50–100 nm and
[97])
gold shell thicknesses of about 4–8 nm are estimated to be
the most effective due to the strong absorption and low
medium on the timescale of ∼100 ps. This fast energy scattering near 800 nm.
conversion and dissipation can be readily used for the
heating of the local environment by using light radiation
with a frequency strongly overlapping with the nanoparticle
SPR absorption band. The intense SPR-enhanced absorp- Bioconjugation and targeting
tion of gold nanoparticles makes the photothermal conver-
sion process highly efficient. The absorption cross section Most laser-based therapeutic methods rely on the use of
of gold nanoparticles [121] is typically four to five orders endoscopes and fiber optic catheters to deliver light
of magnitude stronger than the strongest absorbing Rhoda- specifically to the tumor region and intravenously admin-
Lasers Med Sci (2008) 23:217–228 221

istered photosensitizers/absorbers to achieve selectivity. the antibody has an small net negative charge. The anti-
However, the use of immunotargeting strategies to deliver bodies are thus nonspecifically adsorbed onto gold nano-
the photodynamic or photothermal agents selectively to the particles while still keeping the nanoparticles negatively
diseased cells and tissue offers an efficient route to charged, providing stability in colloidal solution. In recent
achieving selectivity and reducing nonspecific injury to years, Sokolov et al. [136, 137] used this method to bind
healthy cells. Metal, especially gold, nanoparticles allow anti-EGFR to gold nanospheres, which were then used as
easy biofunctionalization, thus making them promising for optical labels to detect the overexpressed EGFR on ovarian
integration with immunotargeting strategies. cancer cells by light scattering imaging using a single
The selectivity of gold nanostructures for photothermal wavelength laser. El-Sayed et al. [139] used the same
cancer therapy has been demonstrated by their functional- method to bind anti-EGFR to gold nanospheres and realized
ization with specific tumor-targeting molecules. Two both diagnostics using dark field microscopy and therapy of
targeting strategies are commonly used. One involves oral cancer cells using a 514 nm argon ion laser [89].
nanoparticles passivated by poly (ethylene) glycol (PEG), Gold nanorods are capped with CTAB molecules, which
and the other employs nanoparticles conjugated to anti- are positively charged [140]. As antibodies are slightly
bodies specific to biomarkers on the diseased cells. PEG is negatively charged at pH=7.4, antibodies can be adsorbed
used to increase the biocompatibility and biostability of onto gold nanorods by electrostatic interactions. However,
nanoparticles and impart them an increased blood retention this causes particle aggregation in solution due to the
time. Citrate-capped gold nanospheres, cetyl trimethylam- neutralization of the surface charge. For the binding of
monium bromide (CTAB)-capped gold nanorods, as well as antibodies to gold nanorods solution, the surface charge is
gold nanoshells have poor stability when they are dispersed reversed by adsorption of a PSS− layer [141, 142]. Then,
in buffer solution due to the aggregating effect of salt ions. the antibodies are adsorbed onto the PSS− layer probably
By capping the nanoparticles with PEG, the biocompatibil- by hydrophobic interactions while still stabilizing the
ity is greatly improved, and nanoparticle aggregation is nanorods by the negative charge on the antibodies [141,
prevented. PEGylated nanospheres and nanorods can be 142]. Figure 3a shows the scheme of the antibody binding
readily made by the conjugation of thiol-functionalized to gold nanorods through a PSS bridging layer.
PEG with the gold nanoparticles [126, 127]. In case of in Another method is the use of chemical binding between
vivo applications, PEGylated nanoparticles are preferen- a functional group on the antibodies and the metal surface
tially accumulated into tumor tissues due to the enhanced of the nanoparticles [143]. The covalent binding of the thiol
permeability and retention effect, known as the “golden group to gold is used very commonly for nanoparticle
standard” for drug design [128–135]. Compared to normal surface bioconjugation. In the work by Liao et al. [126] the
tissue, the blood vessels in tumor tissue are more leaky, and antibodies were bound to nanorods with a long chain
thus, macromolecular or polymeric molecules preferentially succinimidyl 6-[3′-[2-pyridyldithio]-propionamido] (LC-
extravasate into tumor tissue. Due to the decreased SPDP) cross-linker hexanoate (see Fig. 3b). LC-SPDP con-
lymphatics, the tumor tissue retains large molecules for a sists of a pyridildithio group that binds to the gold nanorod
longer time, whereas normal tissue quickly clears out the surface, and an N-hydroxysuccinimide (NHS) ester, which
external particles. This tumor targeting method is called binds to the primary amines in the antibodies.
passive targeting as against the antibody-targeting method For nanoshell bioconjugation [99], antibody molecules
[128–135]. are first bound to a PEG linker orthopyridyl-disulfide
The antibody based targeting is more active, specific and (OPSS)–PEG–NHS through an amidohydroxysuccinimide
efficient. The antibodies are selected to target a specific group (NHS). The antibody-PEG linker complex is then
tumor marker. For instance, anti-epidermal growth factor attached to the nanoshell surface through the sulfur-contain-
receptor (EGFR) antibodies can be employed to target ing OPSS group located at the distal end of the PEG linker.
overexpressed EGFR on oral cancer cells [89, 90, 92] and In addition to antibodies, folic acid can also be used for
cervical cancer cells [136, 137], anti-Her2 for overex- active tumor targeting because cancer cells require exces-
pressed Her2 on breast cancer cells [99] and seprase on sive folic acid, which is a ligand for folate receptors [95,
breast cancer cells [88]. The binding of antibodies to gold 102, 144, 145]. Wei and coworkers [95] have conjugated
nanospheres was first reported in the 1950s when anti- folate ligands with oligoethyleneglycol spacers to gold
bodies were used for the staining of cellular components by nanorods by in situ dithiocarbamate formation. The folate-
electron microscopy [138]. The antibodies are generally conjugated gold nanorods were found to specifically bound
bound to negatively charged citrate-capped gold nano- to the surface of KB cancer cells. Thus, there exist a variety
spheres by nonspecific interactions by adjusting the pH of of surface chemistry techniques that can be utilized for
the colloidal solution to be just above the isoelectric point designing biofunctionalized nanoparticles targeted to the
(pI, zero net charge of the protein) of the antibody such that disease biomarker of choice.
222 Lasers Med Sci (2008) 23:217–228

PPTT by visible lasers spectroscopy, it was found that gold nanoparticles were
preferentially and specifically bound to the cancer cells,
In 1999, Lin et al. [125] first reported the selective PTT while only a heterogeneous nonspecific distribution of the
based on the use of light-absorbing microparticles that nanoparticles was seen over the healthy cells [139]. The
absorb light in the visible region. In 2003, they demon- nanoparticle-labeled cells were then exposed to a CW argon
strated selective PPTT using gold nanoparticle immuno- ion laser at 514 nm [89]. It was found that the malignant
conjugates [84]. Lymphocytes incubated with gold cells required less than half the laser energy to be killed as
nanoparticles conjugated to antibodies and then exposed compared to the benign cells (see Fig. 4, [89]). No
to short laser pulses (565 nm wavelength, 20 ns duration) photothermal destruction was observed for any of the cell
showed cell death with 100 laser pulses at an energy of types without nanoparticle labeling, even at four times the
0.5 J/cm2. The cell death is attributed mainly to the energy required to kill the malignant cells labeled with anti-
cavitation bubble formation around the nanoparticles. By EGFR/gold nanoparticle conjugates. This selective photo-
adjusting the particle number, size, and laser energy, the damage of the cancer cells is clearly attributed to the higher
researchers were able to selectively induce cell death or gold nanoparticle loading on cancer cells due to the
transiently modify cellular functions without causing cell overexpressed EGFR on the cancer cell surface. Higher
destruction. In the same year, Zharov et al. [85] studied the gold nanoparticle labeling results in a consequently higher
threshold and the dynamics of thermal events around the optical density. Thus, a lower laser energy is required to
particles incorporated into K562 cancer cells using nano- raise the temperature above the threshold for destruction, as
second Nd–YAG laser at 532 nm and a photothermal estimated to be in the range of 70–80°C [90]. This method
contrast technique. They found that, at an energy level of can be extended to other types of cancers as well because
2–3 J/cm2, only one or three laser pulses are sufficient to most types of cancer cells have an overexpression of EGFR
damage a cell containing 10–15 particles of 20 nm size, receptors. However, the use of visible light absorbing
whereas at a lower fluence rate of 0.5 J/cm2, at least 50 nanospheres is restricted to skin or near-surface type
pulses and approximately 100 particles are required to cancers due to the inability of visible light to penetrate
produce the same harmful effects on the cells. through skin and tissue.
Recently, El-Sayed and coworkers [89, 90] demonstrated
selective PPTT by using gold nanoparticles with a visible
continuous wave (CW) laser. In these studies, 40 nm gold PPTT by NIR lasers
nanoparticle were conjugated to anti-EGFR antibodies and
then incubated with both human oral cancer cells and For in vivo therapy for tumors under skin and deeply seated
nonmalignant skin cells for 30 min. By using dark field within tissue, NIR light is required because of its deep
light scattering imaging and surface plasmon absorption penetration due to minimal absorption of the hemoglobin

Fig. 3 Antibody conjugation of


gold nanorods. a Gold nanorods
are coated with PSS polyelec-
trolytes and antibodies are
adsorbed onto PSS by nonspe-
cific interactions; b antibodies
are covalently bound to the rod
surface by LC-SPDP crosslinker
(reproduced with permission
from [126])
Lasers Med Sci (2008) 23:217–228 223

Fig. 4 Selective PPTT for can-


cer cells by using anti-EGFR
conjugated gold nanospheres
(reproduced with permission
from [89]). After incubation
with anti-EGFR conjugated gold
nanoparticles, HaCat normal
cells are destroyed at a laser
power threshold of 57 W/cm2,
while HSC and HOC cancer
cells are destroyed at much
lower thresholds of 25 and
19 W/cm2, respectively. The
difference reflects the much
larger density of anti-EGFR
conjugated gold nanospheres on
the surface of the cancer cells
compared to that on the normal
cells due to the selective target-
ing of EGFR overexpressed on
cancer cells

and water molecules in tissues in this spectral region. damaged as compared to the normal cells (20 W/cm2), as
Carbon nanotubes have optical absorbance in the NIR attributed to the selective targeting of the overexpressed
window [146] and, thus, have potential for NIR PTT. Dai EGFR on the cancer cell surface by the anti-EGFR-
and coworkers [102] achieved selective cancer cell conjugated gold nanorods (Fig. 5).
destruction by the functionalization of single-walled carbon Later, Takahashi et al. [93] in Japan achieved cell death
nanotubes with a folate moiety that selectively targets the using phosphatidylcholine-passivated gold nanorods and a
folate receptors on tumor cells, while the receptor-free pulsed Nd–YAG laser at 1,064 nm. Recently, Wei and
normal cells are unaffected. coworkers at Purdue University [95] demonstrated that gold
Gold nanorods and nanoshells have been demonstrated nanorods conjugated to folate ligands can be used for
for selective PPTT using CW NIR lasers mainly by the El- hyperthermic therapy of KB oral cancer cells with a CW
Sayed [92] and Halas groups [96, 99], respectively. By Ti:Sapphire laser. Severe blebbing of cell membranes
using dark-field light scattering imaging, El-Sayed and was observed at laser irradiation with power density as
coworkers found that gold nanorods conjugated to anti- low as 30 J/cm2.
EGFR antibodies were well organized on the surface of The work by Halas and coworkers has shown that gold
cancer cells with relatively higher binding affinity, while nanoshells can be used for PPTT in the NIR region by both
they were randomly distributed nonspecifically on and passive cancer targeting using PEG-conjugated nanoshells
around the normal cells, similar to the case of the gold [96–98] and active targeting using antibody-conjugated
nanospheres [92, 139]. A CW Ti:Sapphire laser with a nanoshells [99]. In the antibody case [99], gold nanoshells
wavelength at 800 nm, overlapping with the SPR absorp- conjugated through PEG linkers to anti-Her2 antibodies
tion wavelength maximum of gold nanorods at 800 nm, were employed for targeting breast cancer cells. A diode
was used for the photoirradiation of the cells labeled with laser at 820 nm was used for photothermal heating (7 min)
the nanorods. It was found that the cancer cells required of the labeled cells (Fig. 6). Only the cancer cells incubated
half the laser energy (10 W/cm2) to be photothermally with the antibody conjugated gold nanoshells were dam-
224 Lasers Med Sci (2008) 23:217–228

Fig. 5 Selective PPTT for can-


cer cells in the NIR region by
using anti-EGFR conjugated
gold nanorods (reproduced with
permission from [92]). After
incubation with anti-EGFR con-
jugated gold nanorods, HaCat
normal cells are destroyed at a
laser power threshold of 20 W/
cm2, while HSC and HOC can-
cerous cells are destroyed at a
much lower threshold of 10 W/
cm2. The difference reflects the
much larger density of gold
nanorods on the surface of the
cancer cells compared to that on
the normal cells

aged under the laser irradiation. In the PEG case [96], the in vivo. For in vivo therapy, the researchers achieved
researchers demonstrated successful PTT both in vitro and successful targeting using the PEGylated gold nanoshells
Fig. 6 Selective PPTT for can-
cer cells by using anti-Her2
antibody conjugated gold nano-
shells (reproduced with permis-
sion from [99]). Only the cells
incubated with anti-Her2 anti-
body conjugated gold nano-
shells are damaged under NIR
irradiation
Lasers Med Sci (2008) 23:217–228 225

injected directly into the tumor region [96] or delivered 9. Seki T, Wakabayashi M, Nakagawa N, Imamura M, Tamai T,
Nishimura A, Yamashiki N, Okamura A, Inoue K (1999)
intravenously [98]. It was shown that NIR light of 820 nm
Percutaneous microwave coagulation therapy for patients with
at 4 W/cm2 caused irreversible tumor tissue damage. Most small hepatocellular carcinoma, Comparison with percutaneous
notably, these studies show that laser dosages required to ethanol injection therapy. Cancer (Philadelphia) 85:1694–1702
induce tissue damage using the plasmonic gold nano- 10. Kremkau FW (1979) Cancer therapy with ultrasound: a historical
review. J Clin Ultrasound 7(4):287–300
structures are 10- to 25-fold lower than those used in
11. Huber P, Debus J, Jenne J, Jochle K, van Kaick G, Lorenz WJ,
studies employing photoabsorbing dyes such as indocya- Wannenmacher M (1996) Therapeutic ultrasound in tumor
nine green dye. therapy. Principles, applications and new development. Radiol-
oge 36(1):64–71
12. Wu F, Chen WZ, Bai J, Zou JZ, Wang ZL, Zhu H, Wang ZB
(2001) Pathological changes in human malignant carcinoma
treated with high-intensity focused ultrasound. Ultrasound Med
Summary Biol 27(8):1099–1106
13. Svaasand LO, Gomer CJ, Morinelli E (1990) On the physical
Plasmonic gold nanostructures thus show great promise for rationale of laser induced hyperthermia. Lasers Med Sci 5:121–
128
the selective PTT for cancer as well as other diseases. We
14. Gould RG (1959) The LASER, light amplification by stimulated
propose the name PPTT for this treatment. It is realized that emission of radiation. The Ann Arbor Conference on Optical
a number of variables need to be further addressed, e.g., Pumping
stability, biocompatibility, and chemical reactions of nano- 15. Maiman TH (1960) Stimulated optical radiation in ruby. Nature
particle bioconjugates in physiological environments, blood 187:493–494
16. Kapany NS, Peppers NA, Zweng HC, Flocks M (1963) Retinal
retention time, tumor extravasation, the fate of the nano- photocoagulation by Lasers. Nature 199:146–149
particles following therapy, etc. We anticipate that the 17. Minton JP, Carlton DM, Dearman JR, McKnight WB, Ketcham
success and promise of the initial use of plasmonic AS (1965) An evaluation of the physical response of malignant
nanoparticles for selective PPTT could be efficiently tumor implants to pulsed laser radiation. Surg Gynaecol Obstet
121:538–544
extended to clinical stage once the optimal parameters of 18. Goldman L (1967) Biomedical aspects of the laser. Springer,
these variables are identified, as is being done through New York
current research studies. 19. Goldman L, Rockwell RJ Jr (1968) Laser Systems and their
applications in medicine and biology. Adv Biomed Eng Med
Phys 1:317–382
20. Mullens F, Jennings B, McClusky L (1968) Incision of tissue by
Acknowledgment We thank the financial support of NCI Center of
carbon dioxide laser. Am Surg 34:717–729
Cancer Nanotechnology Excellence (CCNE) Award (U54CA119338).
21. McKenziei AL (1984) Lasers in surgery and medicine. Phys
Med Biol 29(6):619–641
22. Boulnois JL (1986) Photophysical processes in recent medical
laser developments. Lasers Med Sci 1(1):47–66
23. Sultan RA (1990) Tumour ablation by laser in general surgery.
References
Lasers Med Sci 5:185–193
24. Gibson KF, Kernohan WG (1993) Lasers in medicine. J Med
1. Breasted JH (1930) The Edwin Smith surgical papyrus, vol 1. Eng Technol 17(2):51–57
University of Chicago 25. Brunetaud JM, Mordon S, Maunoury V, Beacco C (1995) Non-
2. Gazelle GS, Goldberg SN, Solbiati L, Livraghi T (2000) Tumor PDT uses of lasers in oncology. Lasers Med Sci 10:3–8
ablation with radio-frequency energy. Radiology (Easton, Pa.) 26. Bown SG (1983) Phototherapy of tumours. World J Surg 7:700–
217:633–646 709
3. GoldBerg SN (2001) Radiofrequency tumor ablation: principles 27. Steger AC, Lees WR, Walmsley K, Bown SG (1989) Interstitial
and techniques. Eur J Ultrasound 13(2):129–147 laser hyperthermia: a new approach to local destruction of
4. Goldberg SN, Dupuy DE (2001) Image-guided radiofrequency tumours. BMJ 299(6695):362–365
tumor ablation: challenges and opportunities—part I. J Vasc 28. Masters A, Bown SG (1990) Interstitial laser hyperthermia in
Interv Radiol 12:1021–1032 tumour therapy. Ann Chir Gynaecol 79(4):244–251
5. Mirza AN, Fornage BD, Sneige N, Kuerer HM, Newman LA, 29. Masters A, Bown SG (1990) Interstitial laser hyperthermia in the
Ames FC, Singletary SE (2001) Radiofrequency ablation of solid treatment of tumours. Lasers Med Sci 5:129–136
tumors. Cancer J 7:95–102 30. Masters A, Bown SG (1992) Interstitial laser hyperthermia. Br J
6. Seegenschmiedt MH, Brady LW, Sauer R (1990) Interstitial Cancer 8(4):242–249
thermoradiotherapy: review on technical and clinical aspects. 31. Siegman AE (1986) Lasers, University Science Books. ISBN 0-
Am J Clin Oncol 13(4):352–363 935702-11-3
7. Urano M, Douple E (1992) Physics of microwave hyperthermia 32. Silfvast WT (1996) Laser fundamentals, Cambridge University
in hyperthermia and oncology, vol 3. Springer, Utrecht, The Press. ISBN 0-521-55617-1
Netherlands, pp 11–98 33. Svelto O (1998) Principles of lasers, 4th edn. (trans. David
8. Sato M, Watanabe Y, Ueda S, Iseki S, Abe Y, Sato N, Kimura S, Hanna). Springer. ISBN 0-306-45748
Okubo K, Onji M (1996) Microwave coagulation therapy for 34. Wilson BC (1986) The physics of photodynamic therapy. Phys
hepatocellular carcinoma. Gastroenterology 110(5):1507–1514 Med Biol 31:327–360
226 Lasers Med Sci (2008) 23:217–228

35. Danniell MD, Hill JS (1991) A history of PDT. Aust N Z J Surg sensitiser Cu [II]-haematoporphyrin: a new approach to cell
61:340–348 photoinactivation. Photochem Photobiol 69:708–712
36. Henderson BW, Dougherty TJ (1992) How does photodynamic 60. Camerin M, Rello S, Villanueva A, Ping X, Kenney ME,
therapy work? Photochem Photobiol 55(1):145–157 Rodgers MAJ, Jori G (2005) Photothermal sensitisation as a
37. Ochsner MJ (1997) Photophysical and photobiological processes novel therapeutic approach for tumours: studies at the cellular
in the photodynamic therapy of tumours. Photochem Photobiol B and animal level. Eur J Cancer 41:1203–1212
39(1):1–18 61. Camerin M, Rodgers MAJ, Kenney ME, Jori G (2005) Photo-
38. Dougherty TJ, Gomer CJ, Henderson BW, Jori G, Kessel D, thermal sensitisation: evidence for the lack of oxygen effect on
Korbelik M, Moan J, Peng Q (1998) Photodynamics therapy. J the photosensitizing activity. Photochem Photobiol Sci 4:251–
Natl Cancer Inst 90(12):889–905 253
39. Dolmans DE, Fukumura D, Jain RK (2003) Photodynamic 62. Welch AJ (1984) The thermal response of laser irradiated tissue.
therapy for cancer. Nat Rev Cancer 3(5):380–387 IEEE J Quantum Electron 20:1471–1481
40. Gold MH (2006) Introduction to photodynamic therapy: early 63. Jacques SL, Prahl SA (1987) Modeling optical and thermal
experience. Dermatol Clin 25(1):1–4 distributions in tissue during laser irradiation. Lasers Surg Med
41. Kim IK, Miller JW (2006) Photodynamic therapy. Intraocular 6:494–503
Drug Delivery 129–141 64. Sturersson C, Andersson-Engels S (1995) A mathematical model
42. Raab O (1900) The effect of fluorescent substances on infusoria. for predicting the temperature distribution in laser-induced
Z Biol 39:524–526 hyperthermia. Experimental evaluation and applications. Phys
43. Jesionek A, Tappeiner VH (1903) Therapeutische Versuche mit Med Biol 40:2037–2052
fluoreszierenden Stoffen. Muench Med Wochneshr 47:2042– 65. He X, Bischof JC (2003) Quantification of temperature and
2044 injury response in thermal therapy and cryosurgery. Crit Rev
44. Hausman W (1911) Die sensibilisierende wirkung deshemato- Biomed Eng 31:355–422
porphyrins. Biochem Z 30:276–286 66. Anderson RR, Parrish JA (1981) Microvasculature can be
45. Figge FHJ, Weiland GS, Manganiello LOJ (1948) Affinity of selectively damaged using dye lasers: a basic theory and
neoplastic embryonic and traumatized tissue for metalloporphyr- experimental evidence in human skin. Lasers Surg Med 1:263–
ins. Proc Soc Exp Biol Med 68:640–641 276
46. Lipson RL, Baldes EJ (1960) The photodynamic properties of a 67. Greenwald J, Rosen S, Anderson, RR, Harrist T, MacFarland F,
particular hematoporphyrin derivative. Arch Dermatol 82:508– Noe J, Parrish JA (1981) Comparative histological studies of the
516 tunable dye (at 577 nm) laser and argon laser: the specific
47. Lipson RL, Baldes EJ (1961) Hematoporphyrin derivative: a vascular effects of the dye laser. J Invest Dermatol 77:305–310
new aid for endoscopic detection of malignant disease. J Thorac 68. Anderson RR, Parrish JA (1983) Selective photothermolysis:
Cardiovasc Surg 42:623–629 precise micro-surgery by selective absorption of pulsed radiation.
48. Moan J (1986) Porphyrin photosensitization and phototherapy. Science 200:524–527
Photochem Photobiol 43:681–690 69. Morelli JG, Tan OT, Garden J, Margolis R, Seki Y, Bol J, Carney
49. Vicente MGH (2001) Porphyrin-based sensitizers in the detec- JM, Anderson ŔR, Furumoto H, Parrish JA (1986) Tunable dye
tion and treatment of cancer: recent progress. Curr Med Chem laser (577 nm) treatment of port wine stains. Lasers Surg Med
Anti-Cancer Agents 1(2):175–194 6:94–99
50. Dougherty TJ (1996) A brief history of clinical photodynamic 70. Polla LL, Margolis RJ, Dover JS, Whitaker D, Murphy GF,
therapy development at Roswell Park cancer institute. J Clin Jacques SL, Anderson RR (1987) Melanosomes are a primary
Laser Med 14:219–221 target of Q-switched ruby laser irradiation in guinea pig skin. J
51. Stilts CE, Nelen MI, Hilmey DG, Davies SR, Gollnick SO, Oseroff Invest Dermatol 89:281–286
AR, Gibson SL, Hilf R, Detty MR (2000) Water-soluble, core- 71. Ara G, Anderson R, Mandel K, Oseroff AR (1988) Absorption
modified porphyrins as novel, longer-wavelength-absorbing sensi- of ns photoradiation of melanosomes generates acoustic waves
tizers for photodynamic therapy. Med Chem 43(12):2403–2410 and induces pigmented melanoma cell toxicity. Photochem
52. Spikes JD (1990) New trends in photobiology (invited review). Photobiol 47:37S–40S
Chlorins as photosensitizers in biology and medicine. J Photo- 72. Chen WR, Adams RL, Heaton E, Dickey DT, Bartels KE,
chem Photobiol B Biol:259–274 Nordquist RE (1995) Chromophore-enhanced laser tumor tissue
53. Rosenthal I (1990) Phthalocyanines as photodynamic sensitizers. photothermal interaction using an 808 nm diode laser. Cancer
Photochem Photobiol 51:351–356 Lett 88:15–19
54. Bonnett R (1995) porphyrin and phthalocyanine photosensitizers 73. Chen WR, Adams RL, Bartels KE, Nordquist RE (1995)
for photodynamic therapy. Chem Soc Rev 24:19–33 Chromophore-enhanced in vivo tumor cell destruction using an
55. Anderson RR, Parrish JA (1983) Selective photothermolysis: SOS-nm diode laser. Cancer Lett 94:125–131
precise microsurgery by selective absorption of pulsed radiation. 74. Jori G, Schindl L, Schindl A, Polo L (1996) Novel approaches
Science 220(4596):524–527 towards a detailed control of the mechanism and efficiency of
56. Parrish JA, Anderson RR, Harrist T, Paul B, Murphy GF (1983) photosensitized processes in vivo. J Photochem Photobiol A
Selective thermal effects with pulsed irradiation from lasers: Chem 102:101–107
from organ to organelle. J Invest Dermatol 80:75s–80s 75. Jori G, Spikes JD (1990) Photothermal sensitizers: possible use
57. Welch AJ (1984) The thermal response of laser-irradiated tissue. in tumour therapy. J Photochem Photobiol B Biol 6:93–101
IEEE J Quantum Electron 12:1471–1475 76. El-Sayed MA (2001) Some interesting properties of metals
58. Jori G, Spikes JD (1990) Photothermal sensitizers: possible use confined in time and nanometer space of different shapes. Acc
in tumor therapy. J Photochem Photobiol B Biol 6:93–101 Chem Res 34:257–264
59. Soncin M, Busetti A, Fusi F, Jori G, Rodgers MAJ (1999) 77. Niemeyer CM (2001) Nanoparticles, proteins, and nucleic acids:
Irradiation of amelanotic melanoma cells with 532 nm high peak Biotechnology meets materials science. Angew Chem Int Ed
power pulsed laser radiation in the presence of the photothermal Engl 40:4128–4158
Lasers Med Sci (2008) 23:217–228 227

78. Daniel MC, Astruc D (2004) Gold nanoparticles: Assembly, 97. Loo CH, Lin A, Hirsch LR, Lee MH, Barton J, Halas NJ, West J,
supramolecular chemistry, quantum-size-related properties, and Drezek RA (2004) Nanoshell-enabled photonics-based imaging
applications toward biology, catalysis, and nanotechnology. and therapy of cancer. Tech Cancer Res Treat 3:33–40
Chem Rev 104:293–346 98. O’Neal DP, Hirsch LR, Halas NJ, Payne JD, West JL (2004)
79. West JL, Halas NJ (2003) Engineered nanomaterials for Photothermal tumor ablation in mice using near infrared
biophotonics applications: improving sensing, imaging, and absorbing nanoshells. Cancer Lett 209:171–176
therapeutics. Annu Rev Biomed Eng 5:285–292 99. Loo C, Lowery A, Halas NJ, West JL, Drezek R (2005)
80. Xia Y, Halas NJ (2005) Shape-controlled synthesis and surface Immunotargeted nanoshells for integrated cancer imaging and
plasmonic properties of metallic nanostructures. MRS Bull therapy. Nano Lett 5:709–711
30:338–348 100. Chen J, Wiley B, Li ZY, Campbell D, Saeki F, Cang H, Au L,
81. Warren CWC, Maxwell DJ, Gao X, Bailey RE, Han M, Nie S Lee J, Li X, Xie Y (2005) Gold nanocages: engineering their
(2002) Luminescent quantum dots for multiplexed biological structure for biomedical applications. Adv Mater 17:2255–2261
detection and imaging. Curr Opin Biotechnol 13:40–46 101. Hu M, Petrova H, Chen J, McLellan JM, Siekkinen AR,
82. Parak WJ, Gerion D, Pellegrino T, Zanchet D, Micheel C, Marquez M, Li X, Xia Y, Hartland GV (2006) Ultrafast laser
Williams SC, Boudreau R, Le Gros MA, Larabell CA, Alivisatos studies of the photothermal properties of gold nanocages. J Phys
AP (2003) Biological applications of colloidal nanocrystals. Chem B 110(4):1520–1524
Nanotechnology 14:R15–R27 102. Shi Kam NW, O’Connell M, Wisdom JA, Dai H (2005) Carbon
83. Katz E, Willner I (2004) Integrated nanoparticle-biomolecule nanotubes as multifunctional biological transporters and near-
hybrid systems: Synthesis, properties, and applications. Angew infrared agents for selective cancer cell destruction. Proc Natl
Chem Int Ed 43:6042–6108 Acad Sci 102(33):11600–11605
84. Pitsillides CM, Joe EK, Wei X, Anderson RR, Lin CP (2003) 103. Faraday M (1857) Experimental relations of gold (and other
Selective cell targeting with light-absorbing microparticles and metals) to light. Philos Trans 147:145–181
nanoparticles. Biophys J 84:4023–4032 104. Turkevich J, Stevenson PC, Hillier J (1951) A study of the
85. Zharov VP, Galitovsky V, Viegas M (2003) Photothermal nucleation and growth processes in the synthesis of colloidal
detection of local thermal effects during selective nanophoto- gold. Discuss Faraday Soc 11:551951
thermolysis. Appl Phys Lett 83(24):4897–4899 105. Kerker M (1969) The scattering of light and other electromag-
86. Zharov VP, Galitovskaya E, Viegas M (2004) Photothermal netic radiation. Academic, New York
guidance for selective photothermolysis with nanoparticles. Proc 106. Papavassiliou GC (1979) Optical properties of small inorganic
SPIE 5319:291–300 and organic metal particles. Prog Solid State Chem 12:185–271
87. Hainfeld JF, Slatkin DN, Smilowitz HM (2004) The use of gold 107. Bohren CF, Huffman DR (1983) Absorption and scattering of
nanoparticles to enhance radiotherapy in mice. Phys Med Biol light by small particles. Wiley, New York
49:N309–N315 108. Kreibig U, Vollmer M (1995) Optical properties of metal
88. Zharov VP, Galitovskaya EN, Johnson C, Kelly T (2005) clusters. Springer, Berlin Heidelberg New York
Synergistic enhancement of selective nanophotothermolysis with 109. Link S, El-Sayed MA (1999) Spectral properties and relaxation
gold nanoclusters: potential for cancer therapy. Lasers Surg Med dynamics of surface plasmon electronic oscillations in gold and
37:219–226 silver nanodots and nanorods. J Phys Chem B 103:8410–8426
89. El-Sayed IH, Huang X, El-Sayed MA (2006) Selective laser 110. Link S, El-Sayed MA (1999) Size and temperature dependence
photo-thermal therapy of epithelial carcinoma using anti-EGFR of the plasmon absorption of colloidal gold nanoparticles. J Phys
antibody conjugated gold nanoparticles. Cancer Lett 239(1):129– Chem B 103:4212–4217
135 111. Link S, El-Sayed MA (2000) Shape and size dependence of
90. Huang X, Jain PK, El-Sayed IH, El-Sayed MA (2006) radiative, non-radiative and photothermal properties of gold
Determination of the minimum temperature required for selec- nanocrystals. Int Rev Phys Chem 19:409–453
tive photothermal destruction of cancer cells using immunotar- 112. S Link, El-Sayed MA (2003) Optical properties and ultrafast
geted gold nanoparticles. Photochem Photobiol 82(2):412–417 dynamics of metallic nanocrystals. Ann Rev Phys Chem 54:331–
91. Khlebtsov B, Zharov V, Melnikov A, Tuchin V, Khlebtsov N 366
(2006) Optical amplification of photothermal therapy with gold 113. Mie G (1908) Contribution to the optics of turbid media,
nanoparticles and nanoclusters. Nanotechnology 17:5167–5179 especially colloidal metal suspensions. Ann Phys 25:377–445
92. Huang X, El-Sayed IH, El-Sayed MA (2006) Cancer cell 114. Gans R (1915) Form of ultramicroscopic particles of silver. Ann
imaging and photothermal therapy in the near-infrared region Phys 47:270–284
by using gold nanorods. J Am Chem Soc 128(6):2115–2120 115. Link S, Mohamed MB, El-Sayed MA (1999) Simulation of the
93. Takahashi H, Niidome T, Nariai A, Niidome Y, Yamada S (2006) optical absorption spectra of gold nanorods as a function of their
Gold nanorod-sensitized cell death: Microscopic observation of aspect ratio and the effect of the medium dielectric constant. J
single living cells irradiated by pulsed near-infrared laser light in Phys Chem B 103:3073–3077
the presence of gold nanorods. Chem Lett 35(5):500–501 116. Link S, El-Sayed MA (2005) Simulation of the optical
94. Takahashi H, Niidome T, Nariai A, Niidome Y, Yamada S (2006) absorption spectra of gold nanorods as a function of their aspect
Photothermal reshaping of gold nanorods prevents further cell ratio and the effect of the medium dielectric constant. J Phys
death. Nanotechnology 17:4431–4435 Chem B 109:10531–10532 (erratum)
95. Huff TB, Tong L, Zhao Y, Hansen MN, Cheng JX, Wei A (2007) 117. Murphy CJ, Sau TK, Gole A, Orendorff CJ, Gao J, Gou L,
Hyperthermic effects of gold nanorods on tumor cells. Nano- Hunyadi S, Li T (2005) Anisotropic metal nanoparticles:
medicine 2(1):125–132 Synthesis, assembly, and optical applications. J Phys Chem B
96. Hirsch LR, Stafford RJ, Bankson JA, Sershen SR, Price RE, 109:13857–13870
Hazle JD, Halas NJ, West JL (2003) Nanoshell-mediated near 118. Nikoobakht B, El-Sayed MA (2003) Preparation and growth
infrared thermal therapy of tumors under MR Guidance. Proc mechanism of gold nanorods (NRs) using seed-mediated growth
Natl Acad Sci 100:13549–13554 method. Chem Mater 15:1957–1962
228 Lasers Med Sci (2008) 23:217–228

119. Oldenburg SJ, Averitt RD, Westcott SL, Halas NJ (1998) 133. McNeil SE (2005) Nanotechnology for the biologist. J Leukoc
Nanoengineering of optical resonances. Chem Phys Lett Biol 78:585–594
288:243–247 134. Kommareddy S, Amiji M (2007) Biodistribution and pharmaco-
120. Prodan EM, Radloff C, Halas NJ, Nordlander P (2003) A kinetic analysis of long-circulating thiolated gelatin nanoparticles
hybridization model for the plasmon response of complex following systemic administration in breast cancer-bearing mice.
nanostructures. Science 302:419–422 J Pharm Sci 96(2):397–407
121. Jain PK, Lee KS, El-Sayed IH, El-Sayed MA (2006) Calculated 135. Pissuwan D, Valenzuela SM, Cortie MB (2006) Therapeutic
absorption and scattering properties of gold nanoparticles of possibilities of plasmonically heated gold nanoparticles. Trends
different size, shape, and composition: applications in biological Biotech 24(2):62–67
imaging and biomedicine. J Phys Chem B 110:7238–7248 136. Sokolov K, Follen M, Aaron J, Pavlova I, Malpica A, Lotan R,
122. Du H, Fuh RA, Li J, Corkan A, Lindsey JS (1998) Photo- Richards-Kortum R (2003) Real-time vital optical imaging of
chemCAD††: a computer-aided design and research tool in precancer using anti–epidermal growth factor receptor antibodies
photochemistry. Photochem Photobiol 68:141–142 conjugated to gold nanoparticles. Cancer Res 63:1999–2004
123. Weissleder R (2001) A clearer vision for in vivo imaging. Nat 137. Sokolov K, Aaron J, Hsu B, Nida D, Gillanwater A, Follen M,
Biotechnol 19:316–317 Macaulay C, Adler-Storthz K, Korgel B, Discour M, Pasqualini
R, Arap W, Lam W, Richartz-Kortum R (2003) Optical systems
124. Lin CP, Kelly MW (1998) Cavitation and acoustic emission
for in vivo molecular imaging of cancer. Technol Cancer Res
around laser-heated microparticles. Appl Phys Lett 72:2800–
Treat 2(6):491–504
2802
138. Hayat MA (1989) Colloidal gold: principles, methods and
125. Lin CP, Kelly MW, Sibayan SAB, Latina MA, Anderson RR
applications, vol 1 edn. Academic, San Diego
(1999) Selective cell killing by microparticle absorption of 139. El-Sayed IH, Huang X, El-Sayed MA (2005) Surface plasmon
pulsed laser radiation. IEEE J Quantum Electron 5:963–968 resonance scattering and absorption of anti-EGFR antibody
126. Liao H, Hafner JH (2005) Gold nanorod bioconjugates. Chem conjugated gold nanoparticles in cancer diagnostics: applications
Mater 17:4636–4641 in oral cancer. Nano Lett 5:829–834
127. Niidome T, Yamagata M, Okamoto Y, Akiyama Y, Takahashi H, 140. Nikoobakht B, El-Sayed MA (2001) Evidence for bilayer
Kawano T, Katayama Y, Niidome Y (2006) PEG-modified gold assembly of cationic surfactants on the surface of gold nanorods.
nanorods with a stealth character for in vivo applications. J Langmuir 17:6368–6374
Control Release 114:343–347 141. Ai H, Fang M, Jones SA, Lvov YM (2002) Electrostatic layer-
128. Maeda H (2001) The enhanced permeability and Retention by-layer nanoassembly on biological microtemplates: platelets.
(EPR) effect in tumor Vasculature: the key role of Tumor- Biomacromolecules 3:560–564
selective macromolecular drug targeting. Adv Enzyme Regul 142. Caruso F, Niikura K, Furlong DN, Okahata Y (1997) Assembly
41:189–207 of alternating polyelectrolyte and protein multilayer films for
129. Maedaa H, Fanga J, Inutsukaa T, Kitamoto Y (2003) Vascular immunosensing. Langmuir 13:3427–3433
permeability enhancement in solid tumor: various factors, 143. Liao H, Hafner JH (2005) Gold nanorod bioconjugates. Chem
mechanisms involved and its implications. Int Immunopharma- Mater 17:4636–4641
col 3:319–328 144. Leamon CP, Low PS (2001) Folate-mediated targeting: from
130. Fang J, Sawa T, Maeda H (2003) Factors and mechanism of diagnostics to drug and gene delivery. Drug Discov Today 6:44–51
“EPR”effect and the enhanced antitumor effects of macromolec- 145. Nayak S, Lee H, Chmielewski J, Lyon LA (2004) Folate-
ular drugs including SMANCS. Adv Exp Med Biol 519:29–49 mediated cell targeting and cytotoxicity using thermoresponsive
131. Paciotti GF, Myer L, Weinreich D, Goia D, Pavel N, McLaughlin microgels. J Am Chem Soc 126:10258–10259
RE, Tamarkin L (2004) Colloidal gold: a novel nanoparticle 146. O’Connell MJ, Bachilo SM, Huffman CB, Moore VC, Strano
vector for tumor directed drug delivery. Drug Deliv 11:169–183 MS, Haroz EH, Rialon, KL, Boul PJ, Noon WH, Kittrell C, Ma
132. Greish K, Sawa T, Fang J, Akaike T, Maeda H (2004) J, Hauge RH, Weisman RB, Smalley RE (2002) Band gap
SMAdoxorubicin, a new polymeric micellar drug for effective fluorescence from individual single-walled carbon nanotubes.
targeting to solid tumors. J Control Release 97:219–230 Science 297:593–596

Anda mungkin juga menyukai