Anda di halaman 1dari 10

Available online at www.sciencedirect.

com

Cancer Letters 269 (2008) 57–66


www.elsevier.com/locate/canlet

Gold nanorod assisted near-infrared plasmonic photothermal


therapy (PPTT) of squamous cell carcinoma in mice
Erin B. Dickerson a,1, Erik C. Dreaden b,1, Xiaohua Huang b,1, Ivan H. El-Sayed c,
Hunghao Chu b, Sujatha Pushpanketh b, John F. McDonald a,
Mostafa A. El-Sayed b,*
a
School of Biology, Ovarian Cancer Institute, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
b
Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology,
901 Atlantic Drive, NW, Atlanta, GA 30332-0400, USA
c
Otolaryngology-Head and Neck Surgery, Comprehensive Cancer Center, University of California at San Francisco, CA 94143, USA

Received 19 December 2007; received in revised form 9 April 2008; accepted 14 April 2008

Abstract

Plasmonic photothermal therapy (PPTT) is a minimally-invasive oncological treatment strategy in which photon energy
is selectively administered and converted into heat sufficient to induce cellular hyperthermia. The present work demon-
strates the feasibility of in vivo PPTT treatment of deep-tissue malignancies using easily-prepared plasmonic gold nanorods
and a small, portable, inexpensive near-infrared (NIR) laser. Dramatic size decreases in squamous cell carcinoma xeno-
grafts were observed for direct (P < 0.0001) and intravenous (P < 0.0008) administration of pegylated gold nanorods in
nu/nu mice. Inhibition of average tumor growth for both delivery methods was observed over a 13-day period, with resorp-
tion of >57% of the directly-injected tumors and 25% of the intravenously-treated tumors.
Published by Elsevier Ltd.

Keywords: Nanotechnology; Photothermal therapy; Nanorod; Hyperthermia; Near-infrared; Polyethylene glycol

1. Introduction [14], and catalytic [15] properties have been


exploited in the areas of cancer imaging [2,16–19],
The application of nanotechnology in medicine diagnostics [6,20,21], and treatment [22–30]. Noble
has been a rapidly growing field in recent years [1– metal nanoparticles provide remarkable opportuni-
8]. A variety of structures with unique structural ties in these applications due to their inherently
[9,10], optical [11,12], electronic [13], magnetic low toxicity [31–33] and strongly enhanced optical
properties associated with localized surface plasmon
resonance (LSPR) [34–36]. The enhanced electro-
*
Corresponding author. Tel.: +1 404 894 0292; fax: +1 404 894 magnetic field surrounding such particles gives rise
0294.
E-mail address: mostafa.el-sayed@chemistry.gatech.edu
to large absorption, Rayleigh (Mie) scattering,
(M.A. El-Sayed). Raman scattering, and two-photon luminescence
1
These authors contributed equally to this work. cross-sections, properties which have been utilized

0304-3835/$ - see front matter Published by Elsevier Ltd.


doi:10.1016/j.canlet.2008.04.026
58 E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66

in photothermal cancer therapy [24–30] (PTT), sur- or nano-scale particles. Photoexcitation of the latter
face enhanced Raman spectroscopic detection [37– two results in non-radiative relaxation and local
39] (SERS), and diagnostic imaging [17–20] heat transfer to the surrounding tumor environ-
applications. ment. In contrast, photodynamic therapy [63–65]
While surgical excision of tumors is a highly (PDT), relies on non-radiative relaxation through
effective method of cancer treatment, curative strat- local formation of cytotoxic singlet oxygen species.
egies for primary tumors located in vital or poorly While PTT and PDT treatments have garnered sig-
accessible tissues remains a challenge. In cases of nificant attention, such methods are inherently lim-
recurrent tumors or those with ill-defined margins, ited by photobleaching effects and absorption cross-
alternative, and multimodal oncological approaches sections several times weaker than those of noble
are employed. The primary [40–42] and adjunctive metal nanoparticles.
[43–46] treatment of cancers by induced hyperther- Recent advances in the field of plasmonics pres-
mia is a well established but burgeoning field of ent new opportunities for both primary and multi-
medical research. Here, temperatures in tumor- modal PTT strategies using noble metal
loaded tissues are elevated to 40–43 °C [47] and nanoparticles. By photo-exciting conduction elec-
above by selective or non-selective application of trons which oscillate at the surfaces of such struc-
microwave, radio, ultrasound, alternating magnetic, tures (surface plasmons), highly efficient local
infrared, or visible radiation. At temperatures heating can be achieved by non-radiative relaxation
greater than 43 °C, protein denaturation and dis- through electron–phonon and subsequent phonon–
ruption of the cellular membrane is known to occur phonon coupling processes [35]. While several mate-
and ablation of tumor tissues has been shown in rials and spherical nanoparticles exhibit surface
numerous cases [42,48,49]. Under mild temperature plasmon resonance in the visible region, opportuni-
increases, clinical studies indicate an acceleration in ties for in vivo plasmonic photothermal therapy [8]
both perfusion and reoxygenation [50,51] of tumor (PPTT) are restricted due to a high degree of
tissues, thereby increasing the efficacy of cytostatic absorption by tissues at visible wavelengths. Such
drug delivery (chemosensitization) and radiotherapy ablative treatments are therefore limited to shallow
(radiosensitization), respectively. In all cases, clini- depths [66]. In contrast, PPTT of deep-tissue malig-
cal studies indicate statistically significant benefits nancies may be accomplished by laser exposure and
to local tumor control and overall survival rates plasmon absorption in the near-infrared region
for primary [40–42] and conjunctive hyperthermia (NIR). Due to minimal attenuation by water and
[52–56]. Although promising, conventional non- hemoglobin at these wavelengths, NIR transmission
invasive hyperthermic strategies are often less selec- [7] in soft tissues may be achieved at depths exceed-
tive than those based-on or used in combination ing 10 cm. By chemically varying the shape or com-
with thermal contrast agents, in many cases, causing position of noble metal nanoparticles [9,21,24,67–
damage to surrounding healthy tissues, as well as 69], surface plasmon absorption can be tuned from
significant discomfort. Moreover, hyperthermic ultraviolet (UV) to infrared (IR) wavelengths. The
treatments using commercially available instru- enhanced non-linear optical properties of spherical
ments are often limited to shallow penetration metal nanoparticles have also been used by our
depths [46] (<3 cm), lower treatment temperatures, group in in vitro near-infrared pulsed laser PPTT
and regions of the body with regular surface compo- by second harmonic generation [29,70,71].
sition. Invasive approaches using microwave anten- The potential uses of gold nanoparticles in near-
nas are highly susceptible to interference, while infrared PPTT have been published using a variety
magnetic particle treatments require large doses. of noble metal nanostructures, including gold nano-
Photothermal therapy [49,57–59] is a minimally- shells [26,48], gold nanorods [8,27,72], and recently,
invasive treatment method in which photon energy gold nanocages [73]. Studies using nanoshell-medi-
is converted to thermal energy sufficient to induce ated PPTT indicate significantly improved local
cellular hyperthermia. Selectivity is achieved by tumor control and survival times in animal models,
focused directional control or invasive [40–42] (fiber while surface plasmon absorption of gold nanocages
optic) positioning of the incident radiation, often has been used in diagnostic imaging and in vitro
pulsed [28–30] or continuous wave [24–28,30,48] therapy [24].
(cw) laser, and is typically accompanied by preferen- One of the simplest and widely used methods to
tial administration of photoactive molecules [60–62] obtain plasmonic nanoparticles involves the seed-
E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66 59

mediated growth of colloidal gold nanorods [68]. 2. Materials and methods


The use of such particles in near-infrared PPTT
is highly attractive due to their rapid synthesis, 2.1. Synthesis and pegylation of gold nanorods
facile bioconjugation, strong absorption cross-sec-
Seed-mediated growth was performed at 25 °C from
tion, and tunable optical extinction. Recent calcu-
freshly prepared aqueous solutions (18 MX) following
lations by discrete dipole approximation (DDA)
methods of Nikoobakht and El-Sayed [68]. Briefly,
show the absorption cross-section of nanorod 2.50 mL of 1.00 mM HAuCl4 (Aldrich, 24459-7) was
structures to be nominally larger than that of added to 5.00 mL of 0.200 M cetyltrimethylammonium
nanocages and more than twice that of nanoshell bromide (CTAB, Aldrich). Six hundred microliters of
structures at their NIR plasmon resonance [73]. ice-cold 10 mM NaBH4 (Aldrich, 480886) was added to
By synthetically varying the aspect ratio of the the stirred solution and allowed to react for several min-
nanorods, longitudinal plasmon absorption can utes, forming the pale brown gold seed solution. Next,
be shifted throughout the visible, NIR, and IR 100.0 mL of 1.00 mM HAuCl4 was added to 100.0 mL
regions [68,74–76]. of 0.200 M CTAB and 4.50 mL of 4.00 mM AgNO3
Our previous work [27] showed that gold nano- (Fischer). Approximately 1.40 mL of 78.8 mM ascorbic
acid (Aldrich, A-7506) was added, followed by gentle
rods conjugated to epithelial growth factor receptor
mixing to form the transparent growth solution. One
antibodies (anti-EGFR) can serve as contrast agents
hundred sixty microliters of the seed solution was added
for in vitro biodiagnostics. Due to overexpression of to the unstirred growth solution and allowed to react for
the EGF receptor on cancer cell surfaces and the 2 h. Nanorods synthesized by this method are approxi-
specificity of antibody binding, malignant cells were mately 12 nm in width and 50 nm length (4.0 aspect
found to require half the laser energy necessary to ratio), with a longitudinal plasmon absorption maximum
destroy normal cells when both were incubated with at 800 nm.
the same concentration of nanorod bioconjugates, a Gold nanorods solutions were centrifuged twice at
key feature of selective PPTT. 20,000g for 15 min and re-dispersed in deionized water
In the present work, the feasibility of in vivo near- to remove excess CTAB molecules. mPEG-SH (Nektar
infrared PPTT is demonstrated using colloidal gold Therapeutics, MW5000) was added to the 1 nM colloi-
dal nanorod solution at a final concentration of 10 mM.
nanorods in an animal model. Subcutaneous squa-
Rods were sonicated overnight and centrifuged at
mous cell carcinoma xenografts were grown in nude
20,000g for 15 min and re-dispersed in deionized water
(nu/nu) mice and particles were selectively delivered to remove non-specifically bound PEG molecules. The
to tumors by both direct and intravenous injection. pegylated gold nanorods were again centrifuged at
Thiolated poly (ethylene) glycol (PEG5000) was 20,000g for 15 min, sterile filtered, and re-dispersed in
covalently bound to the gold nanorod surface to 10 mM phosphate-buffered saline (PBS, Mediatech) to
increase biocompatibility [77–80], suppress the desired optical density at 800 nm. Extinction spectra
immunogenic responses, and to decrease adsorption of the pegylated nanorod saline suspensions showed no
to the negatively charge luminal surface of blood peak shift, broadening, or reduction over a 1-week period
vessels. Near-infrared PPTT was performed prior to injection.
extracorporally using a small, portable, inexpensive,
continuous wave diode laser. Making use of the 2.2. Cell culture and inoculation of mice with tumor cells
enhanced permeability and retention (EPR) effect
[81,82], preferential accumulation of pegylated HSC-3 human squamous carcinoma cells were cul-
gold nanorods in tumor tissues was achieved due tured in DMEM (Mediatech) supplemented with 10% v/
to the high density, extensive extravasation, and v heat-inactivated fetal bovine serum (Invitrogen), 2 mM
inherently defective architecture of the tumor vascu- L-glutamine (Sigma), penicillin (100 U/ml) (Sigma), and

lature, as well the diminished lymphatic clearance streptomycin (100 lg/ml) (Sigma) in a 5% CO2 humidified
from associated interstitial spaces. Significant atmosphere. Female nu/nu mice, 7–8 weeks of age, were
obtained from Taconic (Hudson, NY). Mice were injected
decrease in tumor growth was observed for both
subcutaneously in the flank with 100 lL (3  106) HSC-3
direct tumor injection (P < 0.0001) and intravenous cells suspended in 10 mM PBS. Near-infrared PPTT was
(P < 0.0008) treatments. Inhibition of average performed once tumor burden reached 3 mm in diameter
tumor growth for both delivery methods was (7–9 days). All experiments were conducted with the
observed over a 13-day period, with resorption of approval of the Institutional Animal Care and Use Com-
>57% of the directly-injected tumors and 25% of mittee (IACUC) of the Georgia Institute of Technology
the intravenously-treated tumors. (Atlanta, GA).
60 E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66

2.3. Intratumoral nanoparticle accumulation, temperature, the HSC-3 xenograft model, tumors and vital organs were
and imaging harvested at day 14 for use in separate, ongoing toxicolog-
ical investigations.
Tumor bearing mice were injected with gold nano-
rods (ODk=800 = 120, 100 ll) via the tail vein, and
3. Results and discussion
euthanized at specified time points. Tumor tissues was
excised, fixed in 10% formalin for 24 h, and embedded
3.1. Intratumoral particle accumulation
in paraffin blocks. Blocks were sectioned (5 lm) and
stained using the Silver Enhancer Kit SE-100 (Sigma–
Pegylated gold nanorods were intravenously injected
Aldrich) according to the manufacturer’s instructions.
(tail vein) to assess optimal intratumoral particle accumu-
Incubation time for optimal visualization was deter-
lation. Following injection, HSC-3 tumors were excised at
mined to be 10 min. Silver staining of the tissue sections
varying time intervals, fixed, sectioned, and stained with
were examined using a BX60 Olympus microscope, and
silver to visualize the extent of particle loading. Nanorods
photographed using an Olympus Camedia digital
directly-injected into tumors were used for as a positive
camera.
control (data not shown). Fig. 1a shows a typical histolog-
Thermal transient measurements of HSC-3 tumor
ical section from a HSC-3 tumor injected with 15 lL of
interstitia were obtained using a 33 gauge hypodermic
10 mM PBS (control). Fig. 1b and c illustrate representa-
thermocouple (Omega). The tip of the thermistor was
tive tumor sections following 2 and 6 h of accumulation,
positioned at the tumor center-of-mass and temperatures
respectively. At these time points, no appreciable accumu-
were recorded in 15 s intervals prior to, during, and fol-
lation of particles was observed. In contrast, high particle
lowing NIR exposure for direct (15 lL, ODk=800 = 40,
loading was observed following 24 h of circulation
10 min, 0.9–1.1 W/cm2, 6 mm dia.) and intravenously
(Fig. 1d). Because the highest accumulation, and therefore
(100 lL, ODk=800 = 120, 10 min, 1.7–1.9 W/cm2, 6 mm
PPTT selectivity, was observed at 24 h, this time point
dia.) administered gold nanorods, as well as for compara-
was used for subsequent intravenous near-infrared PPTT
bly exposed sham/NIR treatments (15 lL direct intratu-
treatments.
moral injection of 10 mM PBS).
Transient particle accumulation following direct and
In vivo imaging of pegylated gold nanorod accu-
intravenous administration was monitored by NIR trans-
mulation was monitored by attenuation of near-infra-
mission imaging (Fig. 2). Intensity line-scans of NIR
red transmission (808 nm diode laser, Power
extinction showed marginal diffusion of directly-injected
Technologies) using a custom-built CCD device array.
particles over 3 min, with no subsequent change observed
Control measurements were taken from images
over several hours. Intensity line-scans from NIR trans-
obtained by 15 lL direct intratumoral injection of
10 mM PBS, while directly and intravenously admin-
istered measurements were obtained using previously
mentioned dosages.

2.4. In vivo near-infrared PPTT

Mice were anesthetized with ketamine/xylazine/ace-


promazine. Fifteen microliters of pegylated gold nanorods
(ODk=800 = 40) were directly-injected into the tumor
interstitium or 100 lL of pegylated gold nanorods
(ODk=800 = 120) were intravenously (tail) injected. Con-
trol tumor sites were injected with 15 lL of 10 mM PBS
with no NIR exposure. For direct administration, mouse
tumors were extracorporeally exposed to NIR radiation
(0.9–1.1 W/cm2, 6 mm dia., 10 min) within 2 min of injec-
tion to limit particle diffusion beyond the tumor bound-
aries. For intravenous administration, nanorods were
Fig. 1. Histological sections of silver stained HSC-3 tumor
allowed 24 h circulation to maximize intratumoral particle
xenografts from female nu/nu mice intravenously (tail) injected
accumulation prior to NIR exposure (1.7–1.9 W/cm2,
with (a) 100 lL of 10 mM PBS following 24 h circulation and
6 mm dia., 10 min). Ellipsoidal tumor volume was calcu- pegylated gold nanorods (100 lL, ODk=800 = 120) following (b)
lated as V = (d)2(D)(p/6). Statistical hypothesis testing 2 h, (c) 6 h, and (d) 24 h accumulation. Direct injection of
was performed using Welch’s t-test and non-parametric particles to the tumor interstitium was used as a positive staining
analysis of variance was performed by the Kruskal–Wallis control (data not shown). Arrow indicates staining of red blood
test. Due to the unusually rapid growth rates observed in cells (observed in all tumors).
E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66 61

25

20 PPTT treatment

Δ T (degrees Celsius)
(15 μL OD800nm=40)
15 sham/NIR treatment
(15 μL 10 mM PBS)

10

0 2 4 6 8 10 12 14
time (min)

25

PPTT treatment
20 (100 μL OD800nm=120)

Δ T (degrees Celsius)
sham/NIR treatment
(100 μL 10 mM PBS)
15

10

0
Fig. 2. NIR transmission images of mice prior to PPTT
treatments. Inset shows intensity line-scans of NIR extinction
0 2 4 6 8 10 12 14
at tumor sites for control (j), intravenous (N), and direct (d) time (min)
administration of pegylated gold nanorods. Control mice were
interstitially injected with 15 lL 10 mM PBS alone, while directly Fig. 3. Thermal transient measurements of HSC-3 tumor inters-
administered mice received interstitial injections of 15 lL pegy- titia during (a) direct and (b) intravenous near-infrared PPTT (j)
lated gold nanorods (ODk=800 = 40, 2 min accumulation), and and sham/NIR (d) treatments using pegylated gold nanorods.
intravenously administered mice received 100 lL pegylated gold Direct PPTT treatments were performed by administration of
nanorod (ODk=800 = 120, 24 h accumulation) injections. 15 lL pegylated gold nanorods (ODk=800 = 40, 2 min accumula-
tion) followed by 10 min of 0.9–1.1 W/cm2 NIR laser exposure.
Intravenous PPTT treatments were performed by administration
mission images of HSC-3 tumor sites directly-injected of 100 lL pegylated gold nanorods (ODk=800 = 120, 24 h accu-
with 15 lL of 10 mM PBS show nominal extinction due mulation) followed by 10 min of 1.7–1.9 W/cm2 NIR laser
to increased tissue density, while line-scans obtained fol- exposure. Sham/NIR treatments were performed by administra-
lowing intravenous nanorod delivery at 24 h accumula- tion of 15 lL 10 mM PBS and NIR laser exposure of comparable
tion showed extinction 2.00 times that observed for time and power density. Errors reported as standard deviation.
control sites. Directly-injected tumor sites showed NIR
extinction at 2 min nanorod diffusion more 2.18 times that Heating efficiencies of PPTT treatments (the ratio of
observed by intravenous administration and 4.35 times steady-state temperature change in the presence of plas-
that observed at control sites. monic particles to that NIR exposed in their absence)
were found to be 3.59 ± 0.5 for direct injection and
3.2. Thermal response measurements and nanorod 1.90 ± 0.4 for intravenous injection of pegylated gold
accumulation nanorods. The former value is remarkably similar to that
observed during in vivo near-infrared PPTT treatments
Thermal transient measurements for direct (Fig. 3a) reported by Hirsch et al. [26] by direct injection of gold
and intravenous (Fig. 3b) near-infrared PPTT treatments nanoshells. Observed increases in temperature change
show thermal equilibrium conditions prior to NIR irradi- for sham/NIR treatments using comparable exposure
ation. Rapid heating was observed upon exposure, fol- times and power densities as direct and intravenous
lowed by steady-state equilibrium. Note that >90% of administration conditions correlate well with increases in
the observed temperature increase occurred within the power density. Disparity of direct and intravenous PPTT
first 3 min. Upon removal of NIR exposure, tissues dis- heating efficiency scales proportionately with observed
played expected Newtonian cooling behavior. increases in NIR extinction (4.35 and 2.00 times greater
62 E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66

than control extinction, respectively) and is attributed to 1000 direct


decreased particle loading by intravenous delivery. tail vein
900
Although particle volume and concentration was signifi- control
800
cantly higher for intravenous injections, accumulation is
likely limited by the extent of tumor angiogenesis and 700

uptake by the reticuloendothelial system (RES). Treat- 600


ment selectivity and efficacy was most apparent for direct

Δ V (mm )
3
500
injections; however, both methods showed significantly
400
improved local tumor control.
300

200
3.3. Tumor growth suppression: direct versus intravenous
100
injection
0

Groups of four to six mice were initially used to estab- -100


lish optimal conditions for near-infrared PPTT treatment 0 1 2 3 4 5 6 7 8 9 10 11 12 1
day
of HSC-3 tumor xenografts. Fifteen microliters of pegy-
lated gold nanorods (ODk=800 = 40) were directly admin- Fig. 4. Average change in tumor volume for HSC-3 xenografts
istered at three sites within the tumor interstitium or following near-infrared PPTT treatment by control (), intrave-
100 lL (ODk=800 = 120) were intravenously injected (tail). nous (j), and direct (d) injection of pegylated gold nanorods.
After 2 min, tumors directly-injected with nanorods were Errors for control (n = 10), direct injection (n = 8), and intrave-
subjected to extracorporeal NIR exposure (808 nm, nous injection (n = 7) groups reported as standard error of the
6 mm dia.) and it was determined that 10–15 min of irra- means. Control mice were treated by interstitial injection of 15 lL
10 mM PBS alone, while intravenous PPTT treatments were
diation at 0.9–1.1 W/cm2 was necessary for maximal
performed by administration of 100 lL pegylated gold nanorods
tumor control and minimal damage to surrounding tis-
(ODk=800 = 120, 24 h accumulation) followed by 10 min of 1.7–
sues. After 24 h, tumors intravenously administered with 1.9 W/cm2 NIR laser exposure. Direct PPTT treatments were
nanorods were also subjected to extracorporeal NIR performed by administration of 15 lL pegylated gold nanorods
exposure (808 nm, 6 mm dia.) and it was determined that (ODk=800 = 40, 2 min accumulation) followed by 10 min of 0.9–
10–15 min of irradiation at 1.7–1.9 W/cm2 was necessary 1.1 W/cm2 NIR laser exposure.
for maximal tumor control and minimal damage to sur-
rounding tissues. In addition, no statistically significant
differences in tumor growth were observed for direct P < 0.0008, respectively). Differences in observed efficacy
nanorod injections without NIR exposure, sham/NIR for direct and intravenous treatments gradually increased
exposure alone, and PBS intratumoral injections alone during the experiment, reaching statistical significance at
(P = 0.427 at day 9) (S1). S2 illustrates typically observed the 8% level on day 13. Non-parametric analysis of vari-
tumor resorption and growth inhibition following direct ance for the treated and untreated groups (Table 2) found
injection of pegylated gold nanorods and near-infrared statistically significant differences at the 2% level and
PPTT versus sham/NIR treatments. below for the duration of the experiment. These results
Using previously established treatment conditions, clearly indicate both the selectivity and specificity of
change in tumor volume was recorded over a 13-day per- near-infrared PPTT.
iod for control mice, as well as those treated by intrave- The dramatic changes in observed HSC-3 tumor
nous and direct nanorod injections followed by PPTT growth are attributed to selective hyperemia of malignant
(S3). Here, control mice were subjected to 15 lL direct tissues treated with pegylated gold nanorods by near-
injection of 10 mM PBS to the tumor interstitium, with infrared PPTT. Preferential accumulation of pegylated
no NIR exposure. Average change in tumor volume for gold nanorods within the tumor interstitium occurs due
each group was plotted (Fig. 4) and statistical hypothesis to the EPR effect [81]. Because of their rapid metabolic
testing for differences in average tumor growth was per- rates, tumor cells are regarded as increasingly vulnerable
formed (Table 1). Fig. 4 shows a >96% decrease in aver- to hyperthermic effects [44,72] such as disruption of met-
age tumor growth for directly treated HSC-3 xenografts abolic signaling processes, protein denaturation, and the
and a >74% decrease in average tumor growth for intra- onset of acidosis or apoptosis caused by the production
venously-treated HSC-3 xenografts at day 13 (relative to of heat-shock proteins [83] and other immunostimulants.
control tumors). Moreover, resorption of >57% of the Small increases in local temperature are known to result
directly treated tumors and 25% of the intravenously-trea- in disruption of nuclear and cytoskeletal assemblies and
ted tumors was observed over the monitoring period. indeed, previous [44,84] and recent reports [29,72] indicate
Average tumor growth at day 13 for directly and intra- significant membrane blebbing [85–87] under hyperther-
venously-treated tumors was significantly less than that mic conditions. Under severe conditions, hyperthermic
observed in untreated control groups (P < 0.0001 and damage can result in impaired vasculature supply, endo-
E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66 63

Table 1
P-values for average volume change in HSC-3 tumors following near-infrared PPTT by 808 nm irradiation of pegylated gold nanorods
Day 0 Day 3 Day 6 Day 9 Day 11 Day 13
Direct vs control – 0.0003 0.001 0.0002 0.0001 0.0001
Tail vein vs control – 0.0136 0.0037 0.0005 0.0004 0.0008
Direct vs tail – 0.3415 0.1896 0.1936 0.1205 0.084

Table 2 Appendix A. Supplementary data


Non-parametric analysis of variance for near-infrared PPTT
treatment (2) and control groups
Observed changes in HSC-3 xenograft growth for
Day 0 Day 3 Day 6 Day 9 Day 11 Day 13 intratumoral nanorod injections in female nu/nu
– 0.0024 0.0009 0.0008 0.0001 0.0005 mice without NIR exposure, NIR exposure alone,
and PBS intratumoral injections alone (P = 0.427
at day 9) (S1). S2 illustrates representative tumor
thelial swelling [44], and mictothrombosis associated with resorption and growth inhibition following direct
homeostatic disruption [88]. In vitro, mild hyperthermia
injection of pegylated gold nanorods and near-infra-
has been shown to impede the function of cell surface
red PPTT versus sham/NIR treatments. Change in
receptors, membrane transport, and RNA- and DNA-
polymerization during protein synthesis. Repair of suble- tumor volume for individual mice following estab-
thal cell damage by DNA-polymerase-a and -b, such as lished conditions for near-infrared PPTT treatment
that by incurred during radiotherapy, has also been of HSC-3 xenografts by control (a), intravenous (b),
shown to be inhibited by hyperthermia. While tumor and direct (c) injections of pegylated gold nanorods
growth suppression and resorption is likely a cumulative (S3). Supplementary data associated with this article
result of the previously mentioned effects, it is presumed can be found, in the online version, at doi:10.1016/
here that ablation of the tumor vasculature and localized j.canlet.2008.04.026.
membrane disruption predominates.
Although the mechanism of cellular response in the
present case is yet to be determined, the specificity of References
hyperthermic effects on tumor growth from both direct
[1] M. Ferrari, Cancer nanotechnology: opportunities and
and intravenous near-infrared PPTT treatments is unmis-
challenges, Nat. Rev. Cancer 5 (2005) 161–171.
takable (P < 0.0001 and P < 0.0008, respectively). Inhibi-
[2] X.H. Gao, Y.Y. Cui, R.M. Levenson, L.W.K. Chung,
tion of average tumor growth and minimal damage to S.M. Nie, In vivo cancer targeting and imaging with
surrounding tissues is observed for both methods. Resorp- semiconductor quantum dots, Nat. Biotechnol. 22 (2004)
tion of >57% of the directly-injected tumors and 25% of 969–976.
the intravenously-treated tumors clearly indicates the [3] E. Katz, I. Willner, Integrated nanoparticle–biomolecule
potential curative and adjunctive applications of NIR hybrid systems: synthesis, properties, and applications,
plasmonic photothermal therapy (PPTT) in pre-clinical Angew. Chem. Int. Edit. 43 (2004) 6042–6108.
settings. [4] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating
and target-specific nanoparticles: theory to practice, Phar-
macol. Rev. 53 (2001) 283–318.
[5] C.M. Niemeyer, Nanoparticles, proteins, and nucleic acids:
Acknowledgements
biotechnology meets materials science, Angew. Chem. Int.
Edit. 40 (2001) 4128–4158.
The authors graciously thank Mariam Akhtar [6] N.L. Rosi, C.A. Mirkin, Nanostructures in biodiagnostics,
(G.T.; Department of Biology) for her assistance Chem. Rev. 105 (2005) 1547–1562.
with silver staining studies. Generous support from [7] R. Weissleder, A clearer vision for in vivo imaging, Nat.
Biotechnol. 19 (2001) 316–317.
the Georgia Cancer Coalition (E.B.D.; Distin-
[8] X. Huang, P.K. Jain, I.H. El-Sayed, M.A. El-Sayed, Gold
guished Cancer Scientist Award), the National Can- nanoparticles and nanorods in medicine: from cancer diag-
cer Institute (M.A.E., E.C.D., and X.H.; Center of nostics to photothermal therapy, Nanomedicine 2 (2007) 681–
Cancer Nanotechnology Excellence Award 693.
U54CA119338), the Robinson Family Foundation [9] C. Burda, X.B. Chen, R. Narayanan, M.A. El-Sayed,
Chemistry and properties of nanocrystals of different shapes,
(J.F.M.), Ovarian Cycle (J.F.M.), Golfers against
Chem. Rev. 105 (2005) 1025–1102.
Cancer (J.F.M.), and the US Department of Energy [10] L.M. Liz-Marzan, Tailoring surface plasmons through the
(M.A.E., E.C.D., and X.H.; NO: DE-FG02-97 morphology and assembly of metal nanoparticles, Langmuir
ER14799) is acknowledged. 22 (2006) 32–41.
64 E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66

[11] C. Noguez, Surface plasmons on metal nanoparticles: the [26] L.R. Hirsch, R.J. Stafford, J.A. Bankson, S.R. Sershen, B.
influence of shape and physical environment, J. Phys. Chem. Rivera, R.E. Price, J.D. Hazle, N.J. Halas, J.L. West,
C 111 (2007) 3806–3819. Nanoshell-mediated near-infrared thermal therapy of tumors
[12] M.P. Pileni, Self-assembly of inorganic nanocrystals: fabri- under magnetic resonance guidance, Proc. Natl. Acad. Sci.
cation and collective intrinsic properties, Acc. Chem. Res. 40 USA 100 (2003) 13549–13554.
(2007) 685–693. [27] X. Huang, I.H. El-Sayed, M.A. El-Sayed, Cancer cell
[13] D.M. Adams, L. Brus, C.E.D. Chidsey, S. Creager, C. Creutz, imaging and photothermal therapy in the near-infrared
C.R. Kagan, P.V. Kamat, M. Lieberman, S. Lindsay, R.A. region by using gold nanorods, J. Am. Chem. Soc. 128 (2006)
Marcus, R.M. Metzger, M.E. Michel-Beyerle, J.R. Miller, 2115–2120.
M.D. Newton, D.R. Rolison, O. Sankey, K.S. Schanze, J. [28] C.M. Pitsillides, E.K. Joe, X. Wei, R.R. Anderson, C.P. Lin,
Yardley, X.Y. Zhu, Charge transfer on the nanoscale: current Selective cell targeting with light-absorbing microparticles
status, J. Phys. Chem. B 107 (2003) 6668–6697. and nanoparticles, Biophys. J. 84 (2003) 4023–4032.
[14] L. Thomas, F. Lionti, R. Ballou, D. Gatteschi, R. Sessoli, B. [29] L. Tong, Y. Zhao, T.B. Huff, M.N. Hansen, A. Wei, J.X.
Barbara, Macroscopic quantum tunnelling of magnetization Cheng, Gold nanorods mediate tumor cell death by com-
in a single crystal of nanomagnets, Nature 383 (1996) 145– promising membrane integrity, Adv. Mater. 19 (2007) 3136–
147. 3141.
[15] M.C. Daniel, D. Astruc, Gold nanoparticles: assembly, [30] V.P. Zharov, V. Galitovsky, M. Viegas, Photothermal
supramolecular chemistry, quantum-size-related properties, detection of local thermal effects during selective nanophoto-
and applications toward biology, catalysis, and nanotech- thermolysis, Appl. Phys. Lett. 83 (2003) 4897–4899.
nology, Chem. Rev. 104 (2004) 293–346. [31] E.E. Connor, J. Mwamuka, A. Gole, C.J. Murphy, M.D.
[16] Y.M. Huh, Y.W. Jun, H.T. Song, S. Kim, J.S. Choi, J.H. Wyatt, Gold nanoparticles are taken up by human cells but
Lee, S. Yoon, K.S. Kim, J.S. Shin, J.S. Suh, J. Cheon, In vivo do not cause acute cytotoxicity, Small 1 (2005) 325–327.
magnetic resonance detection of cancer by using multifunc- [32] J.A. Khan, B. Pillai, T.K. Das, Y. Singh, S. Maiti, Molecular
tional magnetic nanocrystals, J. Am. Chem. Soc. 127 (2005) effects of uptake of gold nanoparticles in HeLa cells,
12387–12391. Chembiochem 8 (2007) 1237–1240.
[17] N.J. Durr, T. Larson, D.K. Smith, B.A. Korgel, K. Sokolov, [33] R. Shukla, V. Bansal, M. Chaudhary, A. Basu, R.R.
A. Ben-Yakar, Two-photon luminescence imaging of cancer Bhonde, M. Sastry, Biocompatibility of gold nanoparticles
cells using molecularly targeted gold nanorods, Nano Lett. 7 and their endocytotic fate inside the cellular compartment: a
(2007) 941–945. microscopic overview, Langmuir 21 (2005) 10644–10654.
[18] K. Sokolov, M. Follen, J. Aaron, I. Pavlova, A. Malpica, R. [34] M.A. El-Sayed, Some interesting properties of metals con-
Lotan, R. Richards-Kortum, Real-time vital optical imaging fined in time and nanometer space of different shapes, Acc.
of precancer using anti-epidermal growth factor receptor Chem. Res. 34 (2001) 257–264.
antibodies conjugated to gold nanoparticles, Cancer Res. 63 [35] S. Link, M.A. El-Sayed, Shape and size dependence of
(2003) 1999–2004. radiative, non-radiative and photothermal properties of gold
[19] H. Wang, T.B. Huff, D.A. Zweifel, W. He, P.S. Low, A. Wei, nanocrystals, Int. Rev. Phys. Chem. 19 (2000) 409–453.
J.X. Cheng, In vitro and in vivo two-photon luminescence [36] G. Mie, Contribution to the optics of turbid media,
imaging of single gold nanorods, Proc. Natl. Acad. Sci. USA especially colloidal metal suspensions, Ann. Phys. 25 (1908)
102 (2005) 15752–15756. 377–445.
[20] I.H. El-Sayed, X. Huang, M.A. El-Sayed, Surface plasmon [37] X. Huang, I.H. El-Sayed, W. Qian, M.A. El-Sayed, Cancer
resonance scattering and absorption of anti-EGFR antibody cells assemble and align gold nanorods conjugated to
conjugated gold nanoparticles in cancer diagnostics: appli- antibodies to produce highly enhanced, sharp and polarized
cations in oral cancer, Nano Lett. 5 (2005) 829–834. surface Raman spectra: a potential cancer diagnostic marker,
[21] C. Loo, A. Lowery, N.J. Halas, J.L. West, R. Drezek, Nano Lett. 7 (2007) 1591–1597.
Immunotargeted nanoshells for integrated cancer imaging [38] M. Moskovits, Surface-enhanced spectroscopy, Rev. Mod.
and therapy, Nano Lett. 5 (2005) 709–711. Phys. 57 (1985) 783–826.
[22] M. Johannsen, U. Gneveckow, L. Eckelt, A. Feussner, N. [39] D.A. Stuart, J.M. Yuen, N.S.O. Lyandres, C.R. Yonzon,
Waldofner, R. Scholz, S. Deger, P. Wust, S.A. Loening, A. M.R. Glucksberg, J.T. Walsh, R.P. Van Duyne, In vivo
Jordan, Clinical hyperthermia of prostate cancer using glucose measurement by surface-enhanced Raman spectros-
magnetic nanoparticles: presentation of a new interstitial copy, Anal. Chem. 78 (2006) 7211–7215.
technique, Int. J. Hyperthermia 21 (2005) 637–647. [40] Z. Amin, J.J. Donald, A. Masters, R. Kant, A.C. Steger,
[23] N.W.S. Kam, M. O’connell, J.A. Wisdom, H.J. Dai, Carbon S.G. Bown, W.R. Lees, Hepatic metastases – interstitial laser
nanotubes as multifunctional biological transporters and photocoagulation with real-time US monitoring and
near-infrared agents for selective cancer cell destruction, dynamic CT evaluation of treatment, Radiology 187 (1993)
Proc. Natl. Acad. Sci. USA 102 (2005) 11600–11605. 339–347.
[24] J. Chen, D. Wang, J. Xi, L. Au, A. Siekkinen, A. Warsen, [41] K. Eichler, M.G. Mack, R. Straub, K. Engelmann, S.
Z.Y. Li, H. Zhang, Y. Xia, X. Li, Immuno gold nanocages Zangos, D. Woitaschek, T.J. Vogl, Oligonodular hepatocel-
with tailored optical properties for targeted photothermal lular carcinoma (HCC): MR-guided laser-induced thermo-
destruction of cancer cells, Nano Lett. 7 (2007) 1318–1322. therapy (LITT), Radiology 41 (2001) 915–922.
[25] I.H. El-Sayed, X. Huang, M.A. El-Sayed, Selective laser [42] C.P. Nolsoe, S. Torppedersen, F. Burcharth, T. Horn, S.
photo-thermal therapy of epithelial carcinoma using anti- Pedersen, N.E.H. Christensen, E.S. Olldag, P.H. Andersen,
EGFR antibody conjugated gold nanoparticles, Cancer Lett. S. Karstrup, T. Lorentzen, H.H. Holm, Interstitial hyper-
239 (2006) 129–135. thermia of colorectal liver metastases with a US-guided Nd-
E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66 65

Yag laser with a diffuser tip – a pilot clinical-study, [59] J.M. Brunetaud, S. Mordon, V. Maunoury, C. Beacco, Non-
Radiology 187 (1993) 333–337. PDT uses of lasers in oncology, Lasers Med. Sci. 10 (1995)
[43] M.H. Falk, R.D. Issels, Hyperthermia in oncology, Int. J. 3–8.
Hyperthermia 17 (2001) 1–18. [60] W.R. Chen, R.L. Adams, K.E. Bartels, R.E. Nordquist,
[44] B. Hildebrandt, P. Wust, O. Ahlers, A. Dieing, G. Sreeni- Chromophore-enhanced in-vivo tumor-cell destruction using
vasa, T. Kerner, R. Felix, H. Riess, The cellular and an 808-Nm diode-laser, Cancer Lett. 94 (1995) 125–131.
molecular basis of hyperthermia, Crit. Rev. Oncol. Hematol. [61] W.R. Chen, R.L. Adams, R. Carubelli, R.E. Nordquist,
43 (2002) 33–56. Laser-photosensitizer assisted immunotherapy: a novel
[45] M.R. Horsman, J. Overgaard, Hyperthermia: a potent modality for cancer treatment, Cancer Lett. 115 (1997) 25–
enhancer of radiotherapy, Clin. Oncol. 19 (2007) 418–426. 30.
[46] P. Wust, B. Hildebrandt, G. Sreenivasa, B. Rau, J. Geller- [62] W.R. Chen, R.L. Adams, A.K. Higgins, K.E. Bartels, R.E.
mann, H. Riess, R. Felix, P.M. Schlag, Hyperthermia in Nordquist, Photothermal effects on murine mammary
combined treatment of cancer, Lancet Oncol. 3 (2002) 487– tumors using indocyanine green and an 808-nm diode laser:
497. an in vivo efficacy study, Cancer Lett. 98 (1996) 169–173.
[47] W.C. Dewey, Arrhenius relationships from the molecule and [63] D. Dolmans, D. Fukumura, R.K. Jain, Photodynamic
cell to the clinic, Int. J. Hyperthermia 10 (1994) 457–483. therapy for cancer, Nat. Rev. Cancer 3 (2003) 380–387.
[48] D.P. O’neal, L.R. Hirsch, N.J. Halas, J.D. Payne, J.L. West, [64] T.J. Dougherty, C.J. Gomer, B.W. Henderson, G. Jori, D.
Photothermal tumor ablation in mice using near infrared Kessel, M. Korbelik, J. Moan, Q. Peng, Photodynamic
absorbing nanoshells, Cancer Lett. 209 (2004) 171–176. therapy, J. Nat. Cancer Inst. 90 (1998) 889–905.
[49] R.A. Sultan, Tumour ablation by laser in general surgery, [65] B.W. Henderson, T.J. Dougherty, How does photodynamic
Lasers Med. Sci. 5 (1990) 185–193. therapy work?, J Photochem. Photobiol. 55 (1992) 145–157.
[50] B. Rau, P. Wust, W. Tilly, J. Gellermann, C. Harder, H. [66] J. Elisseeff, K. Anseth, D. Sims, W. Mcintosh, M. Randolph,
Riess, V. Budach, R. Felix, P.M. Schlag, Preoperative R. Langer, Transdermal photopolymerization for minimally
radiochemotherapy in locally advanced or recurrent rectal invasive implantation, Proc. Natl. Acad. Sci. USA 96 (1999)
cancer: regional radiofrequency hyperthermia correlates with 3104–3107.
clinical parameters, Int. J. Radiat. Oncol. Biol. Phys. 48 [67] C.J. Murphy, T.K. San, A.M. Gole, C.J. Orendorff, J.X.
(2000) 381–391. Gao, L. Gou, S.E. Hunyadi, T. Li, Anisotropic metal
[51] C.W. Song, A. Shakil, J.L. Osborn, K. Iwata, Tumour nanoparticles: synthesis, assembly, and optical applications,
oxygenation is increased by hyperthermia at mild tempera- J. Phys. Chem. B 109 (2005) 13857–13870.
tures, Int. J. Hyperthermia 12 (1996) 367–373. [68] B. Nikoobakht, M.A. El-Sayed, Preparation and growth
[52] R.D. Issels, S. Abdel-Rahman, C.M. Wendtner, M.H. Falk, mechanism of gold nanorods (NRs) using seed-mediated
V. Kurze, H. Sauer, U. Aydemir, W. Hiddemann, Neoad- growth method, Chem. Mat. 15 (2003) 1957–1962.
juvant chemotherapy combined with regional hyperthermia [69] B. Wiley, Y. Sun, Y. Xia, Synthesis of silver nanostructures
(RHT) for locally advanced primary or recurrent high-risk with controlled shapes and properties, Acc. Chem. Res. 40
adult soft-tissue sarcomas (STS) of adults: long-term results (2007) 1067–1076.
of a phase II study, Eur. J. Cancer 37 (2001) 1599–1608. [70] X. Huang, P.K. Jain, I.H. El-Sayed, M.A. El-Sayed,
[53] J. Overgaard, D.G. Gonzalez, M. Hulshof, G. Arcangeli, O. Plasmonic photothermal therapy using gold nanoparticles,
Dahl, O. Mella, S.M. Bentzen, Randomized trial of hyper- Lasers Med. Sci. ASAP (2007).
thermia as adjuvant to radiotherapy for recurrent or [71] X. Huang, I.H. El-Sayed, W. Qian, M.A. El-Sayed, The
metastatic malignant-melanoma, Lancet 345 (1995) 540–543. potential use of the enhanced nonlinear properties of gold
[54] B. Rau, P. Wust, P. Hohenberger, J. Loffel, M. Hunerbein, nanospheres in photothermal cancer therapy, Lasers Surg.
C. Below, J. Gellermann, A. Speidel, T. Vogl, H. Riess, R. Med. 39 (2007) 747–753.
Felix, P.M. Schlag, Preoperative hyperthermia combined [72] T.B. Huff, L. Tong, Y. Zhao, M.N. Hansen, J.X. Cheng, A.
with radiochemotherapy in locally advanced rectal cancer – a Wei, Hyperthermic effects of gold nanorods on tumor cells,
phase II clinical trial, Ann. Surg. 227 (1998) 380–389. Nanomedicine 2 (2007) 125–132.
[55] J. Van Der Zee, D.G. Gonzalez, G.C. Van Rhoon, J.D.P. [73] M. Hu, J.Y. Chen, Z.Y. Li, L. Au, G.V. Hartland, X.D. Li,
Van Dijk, W.L.J. Van Putten, A.A.M. Hart, Comparison of M. Marquez, Y.N. Xia, Gold nanostructures: engineering
radiotherapy alone with radiotherapy plus hyperthermia in their plasmonic properties for biomedical applications,
locally advanced pelvic tumours: a prospective, randomised, Chem. Soc. Rev. 35 (2006) 1084–1094.
multicentre trial, Lancet 355 (2000) 1119–1125. [74] R. Gans, Form of ultramicroscopic particles of silver, Ann.
[56] C.C. Vernon, J.W. Hand, S.B. Field, D. Machin, J.B. Phys. 47 (1915) 270–284.
Whaley, J. Vanderzee, W.L.J. Vanputten, G.C. Vanrhoon, [75] A. Gole, C.J. Murphy, Seed-mediated synthesis of gold
J.D.P. Vandijk, D.G. Gonzalez, F.F. Liu, P. Goodman, M. nanorods: role of the size and nature of the seed, Chem. Mat.
Sherar, Radiotherapy with or without hyperthermia in the 16 (2004) 3633–3640.
treatment of superficial localized breast cancer: results from [76] J. Perez-Juste, I. Pastoriza-Santos, L.M. Liz-Marzan, P.
five randomized controlled trials, Int. J. Radiat. Oncol. Biol. Mulvaney, Gold nanorods: synthesis, characterization and
Phys. 35 (1996) 731–744. applications, Coord. Chem. Rev. 249 (2005) 1870–1901.
[57] A.L. Mckenziei, J.A.S. Carruth, Lasers in surgery and [77] J.M. Harris, R.B. Chess, Effect of pegylation on pharma-
medicine, Phys. Med. Biol. 29 (1984) 619–641. ceuticals, Nat. Rev. Drug Discov. 2 (2003) 214–221.
[58] L.O. Svaasand, C.J. Gomer, E.A. Morinelli, On the physical [78] T.B. Huff, M.N. Hansen, Y. Zhao, J.X. Cheng, A. Wei,
rationale of laser induced hyperthermia, Lasers Med. Sci. 5 Controlling the cellular uptake of gold nanorods, Langmuir
(1990) 121–128. 23 (2007) 1596–1599.
66 E.B. Dickerson et al. / Cancer Letters 269 (2008) 57–66

[79] H.W. Liao, J.H. Hafner, Gold nanorod bioconjugates, survival in synchronous G1 Cho cells, J. Cell. Physiol. 126
Chem. Mat. 17 (2005) 4636–4641. (1986) 181–190.
[80] T. Niidome, M. Yamagata, Y. Okamoto, Y. Akiyama, H. [85] M.L. Coleman, E.A. Sahai, M. Yeo, M. Bosch, A. Dewar,
Takahashi, T. Kawano, Y. Katayama, Y. Niidome, PEG- M.F. Olson, Membrane blebbing during apoptosis results
modified gold nanorods with a stealth character for in vivo from caspase-mediated activation of ROCK I, Nat. Cell
applications, J. Control. Release 114 (2006) 343–347. Biol. 3 (2001) 339–345.
[81] H. Maeda, The enhanced permeability and retention (EPR) [86] M. Sebbagh, C. Renvoize, J. Hamelin, N. Riche, J. Berto-
effect in tumor vasculature: the key role of tumor-selective glio, J. Breard, Caspase-3-mediated cleavage of ROCK I
macromolecular drug targeting, Adv. Enzyme Regul. 41 induces MLC phosphorylation and apoptotic membrane
(2001) 189–207. blebbing, Nat. Cell Biol. 3 (2001) 346–352.
[82] R.K. Jain, Transport of molecules in the tumor interstitium: [87] J.C. Mills, N.L. Stone, J. Erhardt, R.N. Pittman,
a review, Cancer Res. 47 (1987) 3039–3051. Apoptotic membrane blebbing is regulated by myosin
[83] G.C. Li, N.F. Mivechi, G. Weitzel, Heat-shock proteins, light chain phosphorylation, J. Cell Biol. 140 (1998)
thermotolerance, and their relevance to clinical hyperther- 627–636.
mia, Int. J. Hyperthermia 11 (1995) 459–488. [88] A. Kowal-Vern, V. Mcgill, J.M. Walenga, R.L. Gamelli,
[84] M.J. Borrelli, R.S.L. Wong, W.C. Dewey, A direct correla- Antithrombin III concentrate in the acute phase of thermal
tion between hyperthermia-induced membrane blebbing and injury, Burns 26 (2000) 97–101.

Anda mungkin juga menyukai