Está en la página 1de 9

www.advmat.

de
www.MaterialsViews.com

Immunological Responses Triggered by Photothermal

COMMUNICATION
Therapy with Carbon Nanotubes in Combination with
Anti-CTLA-4 Therapy to Inhibit Cancer Metastasis

Chao Wang, Ligeng Xu, Chao Liang, Jian Xiang, Rui Peng, and Zhuang Liu*

Despite tremendous effects devoted into cancer research, subcutaneous injection, promising the use of CNTs for cancer
cancer remains one of the major causes of death. The main immunotherapy.[11] Despite the above encouraging results,
reason for such a high mortality from cancer is due to the whether and how CNT-based photothermal therapy would
highly invasive behavior of cancer cells that cause metas- trigger any immulogical response and play any effect in inhib-
tases.[1] Despite improvements in diagnosis and surgical tech- iting tumor metastasis, remain largely unknown.
niques, most deaths from cancer are due to metastasis that is Herein, we report that photothermal ablation of primary
resistant to conventional therapies. Therefore, the next gener- tumors with single-walled carbon nanotubes (SWNTs) in com-
ation of cancer therapy should not only be able to remove or bination with anti-CTLA-4 antibody therapy is able to prevent
destroy the primary tumor, but also to recognize, track down, the development of tumor metastasis in mice. It is found
and destroy any remaining tumor cells at site of distant metas- that polymer-coated SWNTs could not only be used for photo-
tases. Particularly, immunotherapy, in which the patient’s own thermal tumor destruction, which releases tumor-associated
immune system is trained to recognize and destroy tumor cells, antigens, but also act as an immunological adjuvant to greatly
has shown great promise in treating patients with metastatic promote maturation of dendritic cells (DCs) and production of
tumors.[2] anti-tumor cytokines, a unique behavior among several types of
Photothermal therapy (PTT) to treat tumors by light- PTT agents. Furthermore, CTLA-4 blockade applied in SWNT-
induced-heating with the assistant of light-absorbing photo- based photothermal ablation of primary tumors would induce
thermal agents has been wildly studied in recent years.[3] In infiltration of effective T cells while greatly abrogate regula-
the past decade, a large variety of nano-agents including many tory T cells at distant tumors. As the results of such combined
inorganic and organic nanomaterials, all with strong absorb- immunological effects, SWNT-based PTT in combination with
ance in the near-infrared (NIR) tissue transparency window, anti–CTLA-4 therapy is able to inhibit the growth of remaining
have been extensively studied as photothermal agents by our cancer cells in both a subcutaneous tumor model and a lung
and other groups.[4] Among them, carbon nanotubes (CNTs), metastasis model. Our results promise the use of adjuvant
including both single-walled nanotubes (SWNTs) and multi- nanomaterial-based PTT in combination with immunotherapy
walled nanotubes (MWNTs), have been intensively explored for to treat cancer metastasis, a major cause of cancer death.
various biomedical applications including photothermal cancer In our work, SWNTs were modified by a non-covalent
treatment.[5] While being rather effective in killing tumors by approach with a polyethylene glycol (PEG)-grafted amphiphilic
photothermal ablation with lasers,[6] CNTs with appropriate polymer following our well established protocol (Figure 1A).[12]
surface coatings have been found to be not obviously toxic to The obtained PEGylated SWNTs were highly stable in var-
animals and could be gradually excreted from mice over time.[7] ious physiological solutions including PBS, cell medium and
The interactions of CNTs with the immune system, on the serum. Atomic force microscopy (AFM) images revealed
other hand, have also been studied in recent years.[8] Although that those PEGylated SWNTs showed lengths in the range
the results may vary under different experimental conditions, [9] of 100–200 nm (Figure 1B). With a strong absorbance in the
it has been reported that CNTs were able to activate both innate NIR region (Figure S1A, Supporting Information), PEGylated
and adaptive immune responses as potential adjuvants.[10] Par- SWNTs could be rapidly heated up under exposure to an 808-nm
ticularly, Xu and co-workers reported that oxidized MWNTs by NIR laser (Figure S1C–D, Supporting Information), enabling
themselves could induce anti-tumor effect on Babl/c mice upon their use as an effective photothermal agent. PEGylated SWNTs
showed strong resonance Raman scattering (Figure S1B, Sup-
porting Information), indicating that the pristine structure of
C. Wang, Dr. L. Xu, C. Liang, J. Xiang, SWNTs was largely retained after polymer coating.
Prof. R. Peng, Prof. Z. Liu As professional antigen presenting cells (APCs), dendritic
Institute of Functional Nano & cells play pivotal roles in the initiation and regulation of innate
Soft Materials (FUNSOM)
and adaptive immunities. The expression of co-stimulatory
Collaborative Innovation Center of Suzhou
Nano Science and Technology molecules (CD80, CD86), which are the hallmarks of DCs
Soochow University maturation, is a critical factor that determines the quality
Suzhou, Jiangsu 215123, China of the adaptive immunities.[13] Therefore, to understand
E-mail: zliu@suda.edu.cn how our PEGylated SWNTs interact with the immunological
DOI: 10.1002/adma.201402996 system, we firstly studied the effects of PEGylated SWNTs on

Adv. Mater. 2014, © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 1
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

Figure 1. PEGylated SWNTs interact with the immunological system. A) A schematic drawing of our PEGylated SWNTs. B) AFM image of PEGylated
SWNTs. Inset: photograph of PEGylated SWNTs in saline. C) Quantification of CD80+ and CD86+ expression, an indicator of DC maturation, on
bone marrow-derived DCs after in vitro incubation with various concentrations of SWNTs for 12 h. Incubation with PEGylated SWNTs could obviously
enhance the maturation of DCs in vitro by a concentration-dependent manner. Lipopolysaccharides (LPS) was the positive control. D,E) Secretion of
IL-1β (D) and IL-12P70 (E) by DCs after incubation with SWNTs as determined by ELISA. F,G) In vivo DC maturation induced by SWNTs. BALB/c mice
were s.c. injected with SWNTs. Cells in the nearest lymph nodes were examined for the expression of CD11c, CD80, and CD86 by flow-cytometry. H-K)
Cytokine levels in serum from healthy BALB/c mice isolated at 12 h, 72 h and 168 h post SWNT-injection. SWNTs injection enhanced the secretion of
IL6 (H), IL 1β (I), TNF-α (J) and IL 12p70 (K). Data are presented as the mean ± standard errors of the mean (SEM).

DC maturation. For in vitro experiments, bone marrow-derived maturation markers, CD80 and CD86 (Supporting Informa-
DCs separated from BALB/c mice[14] were incubated with var- tion, Figure S3). Remarkably, we found that PEGylated SWNTs
ious concentrations of SWNTs for 12 h. Confocal fluorescence could significantly up-regulate the expression of CD86 & CD80
imaging was conducted for DCs after incubation with fluores- (Supporting Information, Figure S3B–C) and enhance the per-
cently labeled SWNTs, and revealed effective internalization centage of matured DCs (CD80+CD86+ DCs) (Figure 1C). In
of SWNTs by DCs (Supporting Information, Figure S2). Flow addition, we studied the DC stimulation effects of several other
cytometry was then used to determine the expression of DC commonly used PTT agents including PEGlyated graphene

2 wileyonlinelibrary.com © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2014,
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
oxide (GO), PEGlyated gold nanorods (AuNRs), as well as indo- Recent studies have shown that ablative treatments such as
cyanine green (ICG), which, however, all showed much weaker photodynamic therapy, radiofrequency ablation, hyperthermia,
effect to induce DC maturation compared to PEGylated SWNTs high-intensity focused ultrasound (HIFU), as well as cryoabla-
(Figure S4). Our data indicated that SWNTs appeared to be a tion (e.g. ultra-low temperature treatment by liquid nitrogen) of
unique nano-agent to activate DCs, acting as a potential immu- tumors are able to cause tumor-specific immune responses.[15]
nological adjuvant. We thus wonder whether and how SWNT-based photothermal
Next the DC supernatants were harvested to measure inter- ablation of tumors would trigger immunological responses in
leukin 12 (IL-12) and interleukin 1β (IL-1β) by enzyme-linked animal models. Considering the facts that local administra-
immune sorbent assay (ELISA). IL-12 and IL-1β are interleukins tion (e.g. s.c. injection) instead of systemic intravenous (i.v.)
naturally produced by DCs in response to antigenic stimula- injection is usually used for immunotherapy, and the dose
tion. Particularly, IL-12 plays an important role in the activation with local injection of SWNTs could be much lower than that
of cytotoxic activity of NK cells and CD8+ T lymphocytes. It was required with i.v. injection (∼5 mg/kg),[6] in our experiments,
found that the secretion levels of IL-1β and IL-12 by DCs were mice bearing subcutaneous 4T1 murine breast tumors were
both obviously enhanced after SWNTs incubation by a concentra- intratumorally injected with SWNTs (dose = 0.33 mg/kg). After
tion-dependent manner. All these data suggested that DCs could irradiation with an 808-nm NIR laser at 0.5 W/cm2 for 10 min,
be activated by PEGylated SWNTs in vitro. The exact activation the tumor temperature jumped to 53 °C, a temperature high
pathway, however, remains to be understood in future studies. enough to effectively ablate tumor cells (Figure 2A–C). After
To further determine if SWNTs treatment could result in the SWNT-induced photothermal treatment, all tumors on mice
activation of DCs in vivo, healthy mice were subcutaneously were completely eliminated, without showing a single case of
(s.c.) injected with SWNTs (dose = 0.33 mg/kg). Mice were tumor relapse at their original sites after treatment.
sacrificed at 96 h post injection of SWNTs and the maturation The effects of SWNT-based PTT towards DC maturation
status of DCs from the nearest lymph nodes was assessed using and cytokine elevation were then studied. In our experiment,
flow cytometry by co-staining for CD11c, the specific marker of mice were sacrificed at 96 h after SWNT-based PTT to ablate
DCs, together with their maturation markers CD80 and CD86. the 4T1 tumors. The maturation status of DCs within tumor-
It was found that SWNTs could induce the migration of a larger draining lymph nodes (TDLN) was assessed using flow cytom-
number of DCs into lymph nodes and significantly promote the etry. It was found that SWNT-based PTT treatment could
maturation of DCs compared to untreated group (Figure 1F–G greatly promote DC maturation in lymph nodes to a much
& Supporting Information, Figure S5). In addition, we studied greater extent compared to that resulted from SWNT injec-
the in vivo DC stimulation by several other commonly used tion alone without PTT treatment (Figure 2D–H). On the other
PTT agents (GO-PEG, AuNR-PEG, and ICG at the same dose), side, we analyzed various cytokines in the serum of SWNT-
which again showed much weaker effects to induce DC matura- based PTT-treated mice at 12, 72 and 168 hours post-ablation
tion compared to PEGylated SWNTs (Supporting Information, (Figure 2I–M). Both SWNTs alone and SWNT-based PTT were
Figure S6), consistent to our in vitro results. Our data suggest able to increase the secretion of pro-inflammatory cytokines
that SWNTs may as a potential adjuvant which could be recog- IL-1β, IL-12p70, IL-6 and TNF-α. Particularly, the serum level
nized by DCs and lead to DC activation both in vitro and in vivo of TNF-α, which plays an important role in anti-tumor immune
to amplify the subsequent immune responses. responses, was dramatically enhanced after SWNT-treated PTT.
In the following study, we examined multiple cytokines Our data suggest that SWNTs-based PTT is able to initiates a
including IL-1β, IL-12, interleukin 6 (IL-6) (the typical marker significant pro-inflammatory immune response. In situ tumor
of humoral immunity), and tumor necrosis factor α (TNF-α) destruction by SWNTs-based PTT may provide the immune
(typical markers of cellular immunity) in the mouse serum after system with an antigen source for the induction of specific
s.c. injection of SWNTs. It was found that the secretion levels of antitumor immunity as more tumor antigen epitopes may be
IL-1β, IL-12, IL-6, and TNF-α all exhibited apparent elevation exposed after tumor ablation. Therefore, upon contact with
from the untreated control (P value <0.05) within 7 days after SWNTs and tumor cell debris, DCs which are enriched in the
SWNTs injection (Figure 1H–K). While IL-12 could positively skin dermis would be stimulated and migrate to nearby lymph
regulate the activity of natural killer cells, which are the impor- nodes including tumor draining lymph nodes, transit to their
tant composite of innate immunity, TNF-α is a critical mediator mature state up on arrival, activate T lymphocytes and then reg-
for the cellular immunity and plays an important role in tumor ulate subsequent immunities.
immunotherapy. Therefore, we wondered whether SWNTs Inspired by the immune responses induced by SWNT-based
alone would show any tumor inhibition effect on BALB/c mice. PTT, we then wondered whether those effects would be helpful
In our experiment, mice were firstly s.c. injected with SWNTs to prevent tumor metastasis. In our experiment, we inoculated
on their left flanks, and then inoculated with 4T1 murine breast the first tumor by s.c. injection of 4T1 tumor cells on the left
cancer cells on their right flanks. Although the tumor growth flank of each BALB/c mice. One week after injection, when the
in the SWNT-treated group was slightly delayed, such thera- first tumor reached ∼150 mm3, a second tumor was then inocu-
peutic effect was not significant enough particularly at later lated on the right flank. At the following day, the first tumor
time points (Figure S7). Therefore, although PEGylated SWNTs was intratumorally injected with SWNTs and treated with PTT,
upon s.c. injection at the current dose could induce a substan- or simply removed by surgery (Figure 3A). The growth of
tial level of immunological responses including DC activation the secondary tumors, as a mimic of metastatic tumors,
and cytokine secretion, such responses are not strong enough was then monitored afterwards (Figure 3B). Unfortunately,
to inhibit tumor growth by themselves. we found that the immunological responses induced by

Adv. Mater. 2014, © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 3
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

A B
55 oC

25 oC

C 1min 2min 5min 8min 10min

Tempeture (oC)
55

45 Saline PTT
35

25
0 5 10
Time (min)
D F
104 104 104
Untreated SWNTs SWNT based PTT
38.7% 46.3% 67.6%
103 103 103
Counts

CD80 102 102 102

101 101 101

100 100 100


100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
CD11c CD86

E G H
40 40 80

% CD80+ CD86+ DC
% CD11c+ DC

30 30 60
MFI CD80

20 20 40

10 10 20

0 0 0
Untreated SWNTs SWNT Untreated SWNTs SWNT Untreated SWNTs SWNT
baesd PTT baesd PTT baesd PTT

I J K M
20 20 20
Untreated Untreated Untreated Untreated
80
SWNTs SWNTs SWNTs SWNTs
IL12P70 (pg/ml)

15 SWNT-based PTT 15 SWNT-based PTT SWNT-based PTT 15 SWNT-based PTT


TNF-α (pg/ml)

IL1β (pg/ml)

60
IL6 (pg/ml)

10 10 10
40

5 5 20 5

0 0 0 0
12 h 72 h 168 h 12 h 72 h 168 h 12 h 72 h 168 h
12 h 72 h 168 h

Figure 2. SWNT-based PTT induces DC maturation and stimulates the expression of pro-inflammatory cytokines. A) Schematic illustration of the
PTT treatment. B) Representative IR thermal images of tumor-bearing mice exposed to the NIR laser (808 nm, 1 cm2, 10 min) after i.t. injection with
SWNTs. C) Tumor temperature changes for the control mice and SWNT-injected mice under NIR laser irradiation. D-H) DC maturation induced by
SWNT-based PTT to ablate 4T1 tumors on mice. Cells in the TDLN were isolated and stained for CD11c, CD80, and CD86 expressions under flow
cytometry. D) Representative histograms of CD11c expression on DCs analyzed by flow cytometry. E) Qualification of CD11c+ cells according to (D).
F) Representative FACS plots of CD80, CD86 expression gated by CD11c+ cells. G) Corresponding mean fluorescent intensity (MFI) quantification of
CD80. H) Qualification of CD80+ CD86+ cells according to (F). SWNT-based PTT treatment could greatly increase in vivo DC maturation. I-M) Cytokine
levels in sera from mice isolated at 12 h, 72 h and 168 h post SWNT-based PTT. Data are presented as the mean ± SEM.

SWNT-based photothermal ablation of the primary tumors CTLA-4-blocking antibody (anti-CTLA-4), a clinically approved
could not completely inhibit the growth of their secondary therapeutic antibody, is able to target CTLA-4, which is consti-
tumors, whose average growth speed, however, was indeed tutively expressed by regulatory T cells (Tregs) and up-regulated
slower than those on mice with their primary tumors removed after T cell activation.[16] Researchers have found that blockade
by surgery. Additional efforts were thus needed to further of CTLA-4 could inhibit tumor development by abrogating the
enhance the anti-tumor immunological effects induced by immuno-suppressive activity of regulatory T cells (Tregs) and
SWNT-based PTT. augmenting activities of effector T-cells.[16] Clinical trials have
Cytotoxic T lymphocyte antigen-4 (CTLA-4) is a key negative validated the efficacy of anti–CTLA-4 antibody therapy for
regulator of T cell activation. Recent studies have found that the treatment of several types of cancers.[17] In a number of

4 wileyonlinelibrary.com © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2014,
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
A B Surgery
Surgery +Anti CTLA4
SWNT-based PTT

Tumor volume (mm3)


500
SWNT-based PTT+Anti CTLA4
400

300

200

100
D-8 D-1 D0 D1,3,5
0
0 5 10 15
First tumor Second tumor Removal of the Anti-CTLA4
2nd Tumor analysis DAY
Inoculation Inoculation primary tumor by injection
and size
SWNT-based PTT
measurement
or surgery

C D
7.39% 0.0% 15.5% 0.0%
25

CD8+ T Cells (%)


20

15

38.0% 54.6% 39.7% 44.8% 10

Surgery Surgery +Anti–CTLA-4 0


Surgery Surgery + PTT PTT +
7.83% 0.0% 22.9% 0.0%
E Anti-CTLA4 Anti-CTLA4
100
CD4+ T Cells (%)

80
60

13.1% 64.0% 40
7.59% 84.2%
CD8

20
0
SWNT-based PTT SWNT-based PTT + ANTI CTLA4 Surgery Surgery + PTT PTT +
Anti-CTLA4 Anti-CTLA4
CD4
F G 10
Effector to FoxP3 ratio
CD4+ FoxP3+ T Cells(%)

80 CD4Eff/FoxP3 CD8/FoxP3
8

60 6

40
4

20
2

0
Surgery Surgery + PTT PTT + 0
Anti-CTLA4 Anti-CTLA4 Surgery Surgery + PTT PTT +
Anti-CTLA4 Anti-CTLA4

Figure 3. Anti-tumor effect of SWNT-based PTT plus anti–CTLA-4 therapy. A) Schematic illustration of SWNT-based PTT and anti–CTLA-4 combination
therapy. B) Tumor growth curves of different groups of mice after various treatments indicated (7–8 mice per group). Error bars are based on SEM.
C) Quantification of T cells in secondary tumors. Tumor cell suspensions were analyzed by flow cytometry for T cell infiltration (gated on CD3+ T cells).
D,E) Proportion of tumor-infiltrating CD8+ T cells (D) and CD4+ T cells (E) according to (C). F) Proportion of tumor-infiltrating CD4+ FoxP3+ T cells.
G) Ratios of tumor-infiltrating CD8+ T cells and effective CD4+ T cells over regulatory T cells in the secondary tumors upon various treatments to remove
the first tumors. The combination treatment not only increased the ratio of CD8+ T cells to Tregs, but also the ratio of effective CD4+ T cells to Tregs.

previous studies, it has been found that CTLA-4 blockade in The experimental design is shown in Figure 3A. After
combination with cryoablation or hyperthermia could enhance removal of the primary tumors by either photothermal abla-
tumor-specific immune responses and thereby inhibit tumor tion with SWNTs or simply by surgical resection, mice were i.v.
growth at distant sites.[15d,18] Therefore, we next explored injected with three doses of anti-CTLA4 antibody (clone 9H10)
whether CTLA-4 blockage therapy would be able to enhance the (60 µg per mouse for each injection) on day 1, 3, 5. The growth
immunological effects induced by SWNT-based PTT. of secondary tumors as well as the survival of those mice after

Adv. Mater. 2014, © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 5
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

various treatments were monitored (Figure 3B). It was found immunities since there existing IgG Fc fragment receptors on
that anti-CTLA4 treatment could only slightly delay the growth APCs.[21]
of secondary tumors on mice when their primary tumors To further assess the potency of this combination therapy, we
were removed by surgery. Remarkably, for mice with their pri- tested it in a lung metastasis tumor model by challenging mice
mary tumors ablated by SWNT-based PTT, CTLA4 blockage with i.v. injection of 4T1 cells (1*105 cells per mouse), which
could greatly inhibit the development of secondary tumors afterwards would spread into various mouse organs particu-
(Figure 3B, Figure S8&9). This result indicated that although larly into the lung. The lung metastasis was introduced one day
SWNT-based PTT alone could only partially delay the growth before SWNT-based PTT or surgery treatment to remove the
of a second tumor challenge, its anti-tumor efficacy would be primary subcutaneous tumors. In the following days, mice
greatly promoted when combined with the CTLA-4 blockade were i.v. injected with anti-CTLA-4 antibody (60 µg per mouse
strategy, leading to effective rejection of secondary tumors, at day 1, 3, 5) (Figure 4A). On day 10, lungs were harvested
which mimic tumor metastasis. for analysis. Compared to mice with only surgical resection
To investigate the underlying mechanism of tumor inhibi- of the primary tumors, those after SWNT-based PTT treat-
tion by SWNT-based PTT and anti–CTLA-4 combination treat- ment together with anti-CTLA4 therapy showed remarkably
ment, immune cells in secondary tumors were then analyzed. reduced lung metastasis, as evidenced by photos of India-ink
T-cell infiltration in the secondary tumor was studied 10 days stained whole lungs and Hematoxylin and Eosin (H&E) stained
after their implantation by flow cytometry. CD8+ killer T cells, lung slices (Figure 4B&C). With the combination therapy, the
also known as cytotoxic T (Tc) cells, could kills cancer cells average number of metastasis sites in lungs was dramatically
by releasing cytotoxins perforin, granzymes and granulysin decreased from ∼30 per mouse (surgery) to ∼1 per mouse
that eventually lead to apoptosis of targeted cancer cells. The (SWNT-based PTT plus anti-CTLA4 therapy) (Figure 4D). A
percentage of CD8+ T cells in the secondary tumor after the marked difference was also seen for the average lung weights
combined SWNT-based PTT plus anti-CTLA-4 treatment was between these two groups (Figure 4E).
3-fold of that in the surgery treatment or PTT treatment alone Next we studied the survival of mice treated with our com-
groups, and 1.5-fold compared to that in the surgery plus bination therapy. 1*105 4T1 cells were i.v. introduced one day
anti–CTLA4 treatment group (Figure 3C–D). On the other before SWNT-based PTT or surgery treatment to remove the
hand, the percentage of CD4+ helper T (Th) lymphocytes, primary tumor. Every group had 7–8 mice. It was found that
which play critical roles in the regulation of adaptive immuni- 57% of mice survived in 50 days after combination therapy
ties, showed a significant increase in the secondary tumors of (SWNT-based PTT plus anti-CTLA4 therapy). In contrast, only
mice after their primary tumors were ablated by SWNT-based 25% mice could survive with CTLA-4 blockade plus surgery
PTT (Figure 3E). However, for SWNT-based PTT alone, treatment in 50 days, and none of mice could survive with sur-
most of increased CD4+ T cells were found to be regulatory gery or SWNT-based PTT alone treatment without the injec-
T cells, which are able to hamper effective anti-tumor immune tion of anti-CTLA4 (Figure 4F). Our results thus demonstrate
responses (Figure 3F & Figure S10). In marked contrast, that SWNT-based photothermal ablation of the primary tumor
CTLA-4 blockade therapy could greatly abrogate the activity of in combination with anti-CTLA4 treatment would be able to
Tregs and reduce the percentage of Tregs in secondary tumors induce strong anti-tumor immunological responses to inhibit
after SWNT-based photothermal ablation of primary tumors. lung metastasis, showing remarkably therapeutic advantage in
As the results, the combination treatment not only increased prolonging the animal survival.
the ratio of CD8+ T cells to Tregs, but also increased effective At last, to see if the immunological effect of SWNT-based
CD4+ T cells to Tregs (Figure 3G). Both effects are of great PTT is a general behavior towards other types of cancer cell
importance to enhance anti-tumor immunological responses. lines, we have conducted in vitro experiments with another cell
Beside tumor-infiltration T cells, CD20+ tumor-infiltrating line, murine B16 musculus skin melanoma. It was found that
B cells have also been reported to be strongly associated residues of both 4T1 cells and B16 cells after SWNT-induced
with favorable outcomes in immunotherapy of breast, lung, photothermal ablation could strongly activate DC maturation
ovarian and cervical cancers.[19] Nelson and colleagues dem- in an in vitro Transwell system experiment to mimic in vivo
onstrated that CD20+ tumor-infiltrating B lymphocytes (TILs) tumor model (Supporting Information, Figure S12). Although
were able to work together with CD8+ T cells to promote the further in vivo validations are still required, it is likely that the
patient survival through serving as APCs and/or generating immunological responses triggered by photothermal tumor
cytokines to modulate helper T cells polarizing towards Th1 ablation with SWNTs may be applied for different tumor
or Th2 functional phenotype.[20] Therefore, CD20+ TILs were types.
also analyzed in secondary tumors after various treatments. Generally speaking, the underlying mechanisms of the com-
Interestingly, we found that SWNT-based PTT increased the bination therapy with ideal inhibition activities on tumor growth
number of infiltrating B cells (CD20+CD44+ cells) within the and metastasis may greatly correlate with several parameters as
tumor (Figure S11A–B). Meanwhile, it was observed that the follows (Figure 5). (1) SWNTs may act as an ‘adjuvant’ being
level of IgG in the serum of combination treatment group recognized by DCs and leading to DC activation/maturation in
was apparently higher than that in the groups after surgical vivo to amplify the subsequent immune responses. (2) SWNT-
resection plus anti-CTLA4 treatment, as well as surgical resec- based PTT provides tumor-associated antigens for APCs such
tion alone (Figure S11C). This may be greatly useful for APCs as DCs to trigger anti-tumor immune responses. (3) Anti-
including B cells to take up, process and present the tumor- CTLA4 blockade strategy effectively hampers the immune-
associated antigen to T cells and then to initiate the adaptive suppression activity of Tregs and significant increases the ratios

6 wileyonlinelibrary.com © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2014,
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
A

D-8 D-1 D0 D1,3,5

Removal of the
First tumor Lung metastasis primary tumor by Anti-CTLA4 Lung metastasis
Inoculation induction SWNT-based PTT injection Quantification
or surgery

B Surgery C

PTT + Anti-CTLA4

D E F
50 500
Percent survival

100
NO. of lung metastases

Lung Mass (mg)

40 400

50
30 300

20 200
0
0 10 20 30 40 50
10 100 DAY
SWNT-based PTT + Anti CTL4
0 0 SWNT-based PTT
Surgery PTT + Surgery PTT + Surgery + Anti CTLA4
Anti-CTLA4 Anti-CTLA4 Surgery

Figure 4. Lung metastasis inhibition by SWNT-based PTT and anti–CTLA-4 combination therapy. A) Schematic illustration of SWNT-based PTT and
anti–CTLA-4 combination therapy for treatment of a lung metastasis tumor model. B,C) Representative lung photographs (B) and H&E-stained lung
slices (C) collected from mice post different treatments indicated. D,E) Quantification of lung metastasis nodules (D) and lung masses (E) for mice
after different treatments. Compared to mice with only surgical resection of the primary tumors, those after SWNT-based PTT together with anti-CTLA4
therapy showed remarkably reduced lung metastasis. F) The survival curves of mice in 50 days after various treatments indicated. SWNT-based pho-
tothermal ablation of the primary tumor in combination with anti-CTLA4 treatment would be able to induce strong anti-tumor immunological effects
to inhibit lung metastasis.

between CD4+ Th cells to Treg as well as CD8+ Tc cells to Tregs tumor cells inside the body, useful for the treatment of meta-
in secondary tumors. This is greatly helpful for the induc- static tumors.
tion of anti-tumor immunities especially cellular immunity. In summary, we have shown here that SWNT-based PTT
(4) The CD20+ tumor-infiltrating B cells may also serve as APCs together with anti–CTLA-4 therapy could modulate the adaptive
to effectively take up, process and present the tumor-associated immune responses especially cellular immunity for the treat-
antigens for the induction of CTL-mediated cellular immunity ment of metastatic cancer. While being a powerful photothermal
for tumor immunotherapy. Those immune responses taken agent to effectively burn the tumor, PEGylated SWNTs could
together would result in the effective rejection of remaining also act as a potential adjuvant to stimulate DC maturation

Adv. Mater. 2014, © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 7
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

Tumor antigen Dendritic cells Tumor-draining lymph nodes (TDLN)

maturation Th cells
CD4+ Th cells

SWNTs
Tumor antigens
Danger signals (e.g. ROS, HSP…) Tc cells

Primary tumor Inflammatory response


treated with
SWNT-based PTT IL-1β Antigen presentation to naïve T cell
IL-6
IL-12
B cells TNF-α
……
Anti-CTLA4 mAb

e.g. NK cells

Tumor specific antibody


CTLA4
Treg cells
CD8+ Tc cells CD4+ Th cells
Metastasis tumor Tumor specific T cells CTLA-4 blockade

Figure 5. The hypothesized mechanism of anti-tumor immune responses induced by SWNT-based PTT in combination with the anti-CTLA-4
therapy.

and cytokine secretion. Such behaviors are not found or to a Acknowledgements


much weaker extend for several other types of widely studied This work was partially supported by the National Natural Science
photothermal agents including GO, AuNRs and ICG. After Foundation of China (51222203, 51132006), the National “973” Program
SWNT-based photothermal ablation of the primary tumor, the of China (2011CB911002, 2012CB932601), a Juangsu Natural Science
induced immunological responses combined with anti-CTLA4 Fund for Distinguished Young Scholars, the Jiangsu Key Laboratory
treatment are able to inhibit the challenge of a secondary tumor for Carbon-Based Functional Materials, and a Project Funded by the
or even lung metastasis. Our work indicates that compared Priority Academic Program Development of Jiangsu Higher Education
Institutions. The authors thank Prof. Hongjie Dai for his helpful
with surgery resection, the most commonly used cancer treat-
suggestions to this work.
ment strategy, nanomaterials-based photothermal treatment
of cancer may be of advantageous in terms of triggering the Received: July 7, 2014
adaptive immunological responses against remaining tumor Revised: August 29, 2014
cells in the body. This work is the first demonstration that Published online:
photothermal therapy induced by an adjuvant-like nano-agent
in combination with immunotherapy is able to inhibit cancer
metastasis. The mechanisms of the adjuvant-like behaviors
of PEGylated SWNTs still need further careful investigations. [1] a) C. L. Chaffer, R. A. Weinberg, Science 2011, 331, 1559;
Besides SWNTs, which exhibit unique multiple functions in b) J. A. Joyce, J. W. Pollard, Nat. Rev. Cancer 2009, 9, 239.
[2] a) C. M. Britten, H. Singh-Jasuja, B. Flamion, A. Hoos, C. Huber,
this work, it also remains to be explored how other types of
K. J. Kallen, S. N. Khleif, S. Kreiter, M. Nielsen, H. G. Rammensee,
photothermal nano-agents would behave in such combina- U. Sahin, T. Hinz, U. Kalinke, Nat. Biotechnol. 2013, 31,
tion therapy. Nevertheless, the combination between PTT 880; b) H. Ledford, Nature 2014, 508, 24; c) I. Melero, S. Hervas-
nanomedicine-treatment together with antibody-based immu- Stubbs, M. Glennie, D. M. Pardoll, L. Chen, Nat. Rev. Cancer 2007,
notherapy may be a novel cancer therapeutic strategy, which 7, 95.
not only is able to destroy the primary tumor, but also to inhibit [3] a) M. P. Melancon, M. Zhou, C. Li, Acc. Chem. Res. 2011, 44,
cancer metastasis at distant sites. It is thus likely that photo- 947; b) U. Lindner, R. A. Weersink, M. A. Haider, M. R. Gertner,
thermal nano-therapy is not just burning of tumors; instead it S. R. H. Davidson, M. Atri, B. C. Wilson, A. Fenster, J. Trachtenberg,
may offer clinically valuable therapeutic advantages over sur- J. Urology 2009, 182, 1371; c) E. C. Dreaden, M. A. Mackey,
gery in cancer treatment. X. Huang, B. Kang, M. A. El-Sayed, Chem. Soc. Rev. 2011, 40, 3391;
d) S. Lal, S. E. Clare, N. J. Halas, Acc. Chem. Res. 2008, 41, 1842;
e) O. Akhavan, E. Ghaderi, Small 2013, 9, 3593.
[4] a) Q. W. Tian, M. H. Tang, Y. G. Sun, R. J. Zou, Z. G. Chen,
M. F. Zhu, S. P. Yang, J. L. Wang, J. H. Wang, J. Q. Hu, Adv. Mater.
Supporting Information 2011, 23, 3542; b) K. Yang, L. Z. Feng, X. Z. Shi, Z. Liu, Chem.
Supporting Information is available from the Wiley Online Library or Soc. Rev. 2013, 42, 530; c) Z. J. Zhang, L. M. Wang, J. Wang,
from the author. X. M. Jiang, X. H. Li, Z. J. Hu, Y. H. Ji, X. C. Wu, C. Y. Chen,

8 wileyonlinelibrary.com © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2014,
DOI: 10.1002/adma.201402996
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
Adv. Mater. 2012, 24, 1418; d) J. F. Lovell, C. S. Jin, E. Huynh, [10] a) J. Palomaki, E. Valimaki, J. Sund, M. Vippola, P. A. Clausen,
H. Jin, C. Kim, J. L. Rubinstein, W. C. Chan, W. Cao, L. V. Wang, |K. A. Jensen, K. Savolainen, S. Matikainen, H. Alenius, ACS Nano
G. Zheng, Nat. Mater. 2011, 10, 324; e) M. S. Yavuz, Y. Cheng, 2011, 5, 6861; b) E. Meunier, A. Coste, D. Olagnier, H. Authier,
J. Chen, C. M. Cobley, Q. Zhang, M. Rycenga, J. Xie, C. Kim, K. H. Song, L. Lefèvre, C. Dardenne, J. Bernad, M. Béraud, E. Flahaut, B. Pipy,
A. G. Schwartz, Nat. Mater. 2009, 8, 935; f) O. Akhavan, Nanomed-Nanotechnol. 2012, 8, 987.
A. Meidanchi, E. Ghaderi, S. Khoei, J. Mater. Chem. B 2014, [11] M. Yang, J. Meng, X. Cheng, J. Lei, H. Guo, W. Zhang, H. Kong,
2, 3306. H. Xu, Theranostics 2012, 2, 258.
[5] a) H. Gong, R. Peng, Z. Liu, Adv. Drug Deliver. Rev. 2013, 65, [12] C. Wang, X. X. Ma, S. Q. Ye, L. Cheng, K. Yang, L. Guo, C. H. Li,
1951; b) Z. Liu, K. Chen, C. Davis, S. Sherlock, Q. Z. Cao, Y. G. Li, Z. Liu, Adv. Funct. Mater. 2012, 22, 2363.
X. Y. Chen, H. J. Dai, Cancer Res. 2008, 68, 6652; c) K. Welsher, Z. Liu, [13] C. A. Janeway, K. Bottomly, Cell 1994, 76, 275.
S. P. Sherlock, J. T. Robinson, Z. Chen, D. Daranciang, H. J. Dai, [14] M. B. Lutz, N. Kukutsch, A. L. J. Ogilvie, S. Rossner, F. Koch,
Nat. Nanotechnol. 2009, 4, 773; d) J. W. Kim, E. I. Galanzha, N. Romani, G. Schuler, J. Immunol. Methods 1999, 223, 77.
E. V. Shashkov, H. M. Moon, V. P. Zharov, Nat. Nanotechnol. 2009, [15] a) J. P. M. Almeida, R. A. Drezek, A. E. Foster, Immunotherapy 2014,
4, 688; e) Z. Liu, S. Tabakman, K. Welsher, H. J. Dai, Nano Res. 6, 109; b) S. R. Gameiro, J. P. Higgins, M. R. Dreher, D. L. Woods,
2009, 2, 85; f) T. R. Fadel, E. R. Steenblock, E. Stern, N. Li, X. Wang, G. Reddy, B. J. Wood, C. Guha, J. W. Hodge, Plos One 2013, 8,
G. L. Haller, L. D. Pfefferle, T. M. Fahmy, Nano Lett. 2008, 8, 2070; e70417; c) Q. Liu, B. Zhai, W. Yang, L.-X. Yu, W. Dong, Y.-Q. He,
g) K. Kostarelos, A. Bianco, M. Prato, Nat. Nanotechnol. 2009, L. Chen, L. Tang, Y. Lin, D.-D. Huang, Mol. Ther. 2009, 17, 2049;
4, 627; h) B. R. Smith, E. E. Ghosn, H. Rallapalli, J. A. Prescher, d) R. Waitz, S. B. Solomon, E. N. Petre, A. E. Trumble, M. Fassò,
T. Larson, L. A. Herzenberg, S. S. Gambhir, Nat. Nanotechnol. 2014, L. Norton, J. P. Allison, Cancer Res. 2012, 72, 430; e) J. E. Kennedy,
9, 481; i) Z. Liu, W. Cai, L. He, N. Nakayama, K. Chen, X. Sun, Nat. Rev. Cancer 2005, 5, 321; f) K. F. Chu, D. E. Dupuy, Nat. Rev.
X. Chen, H. Dai, Nat. Nanotechnol. 2007, 2, 47. Cancer 2014, 14, 199.
[6] a) P. Chakravarty, R. Marches, N. S. Zimmerman, A. D. Swafford, [16] a) F. S. Hodi, Clin. Cancer Res. 2007, 13, 5238; b) T. Pentcheva-Hoang,
P. Bajaj, I. H. Musselman, P. Pantano, R. K. Draper, E. S. Vitetta, E. Corse, J. P. Allison, Immunol. Rev. 2009, 229, 67;
Proc. Natl. Acad. Sci. USA 2008, 105, 8697; b) X. Liu, H. Tao, c) J. F. Grosso, M. N. Jure-Kunkel, Cancer Immun. 2013, 13, 5.
K. Yang, S. Zhang, S. T. Lee, Z. Liu, Biomaterials 2011, 32, 144; [17] T. Yervoy, Princeton, NJ: Bristol-Myers Squibb Company 2011.
c) H. K. Moon, S. H. Lee, H. C. Choi, ACS Nano 2009, 3, 3707; [18] M. H. den Brok, R. P. Sutmuller, R. van der Voort, E. J. Bennink,
d) J. T. Robinson, K. Welsher, S. M. Tabakman, S. P. Sherlock, C. G. Figdor, T. J. Ruers, G. J. Adema, Cancer Res. 2004, 64, 4024.
H. Wang, R. Luong, H. Dai, Nano Res. 2010, 3, 779. [19] T. W. Kuijpers, R. J. Bende, P. A. Baars, A. Grummels, I. A. M. Derks,
[7] a) R. Singh, D. Pantarotto, L. Lacerda, G. Pastorin, C. Klumpp, K. M. Dolman, T. Beaumont, T. F. Tedder, C. J. M. van Noesel,
M. Prato, A. Bianco, K. Kostarelos, Proc. Natl. Acad. Sci. USA 2006, E. Eldering, R. A. W. van Lier, J. Clin. Invest. 2010, 120, 214.
103, 3357; b) Z. Liu, C. Davis, W. Cai, L. He, X. Chen, H. Dai, Proc. [20] a) J. S. Nielsen, B. H. Nelson, Oncoimmunology 2012, 1, 1623;
Natl. Acad. Sci. USA 2008, 105, 1410; c) A. De La Zerda, C. Zavaleta, b) J. S. Nielsen, R. A. Sahota, K. Milne, S. E. Kost, N. J. Nesslinger,
S. Keren, S. Vaithilingam, S. Bodapati, Z. Liu, J. Levi, B. R. Smith, P. H. Watson, B. H. Nelson, Clin. Cancer Res. 2012, 18, 3281.
T. J. Ma, O. Oralkan, Z. Cheng, X. Y. Chen, H. J. Dai, [21] a) S. Amigorena, C. Bonnerot, Semin Immunol. 1999, 11, 385;
B. T. Khuri-Yakub, S. S. Gambhir, Nat. Nanotechnol. 2008, 3, 557. b) C. Scuoppo, C. Miething, L. Lindqvist, J. Reyes, C. Ruse,
[8] a) H. Dumortier, Adv. Drug Deliver. Rev. 2013, 65, 2120; I. Appelmann, S. Yoon, A. Krasnitz, J. Teruya-Feldstein, D. Pappin,
b) T. R. Fadel, T. M. Fahmy, Trends Biotechnol. 2014, 32, 198. J. Pelletier, S. W. Lowe, Nature 2012, 487, 244; c) K. Serre, P. Machy,
[9] A. J. Andersen, P. P. Wibroe, S. M. Moghimi, Adv. Drug Deliver. Rev. J. C. Grivel, G. Jolly, N. Brun, J. Barbet, L. Leserman, J. Immunology
2012, 64, 1700. 1998, 161, 6059.

Adv. Mater. 2014, © 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 9
DOI: 10.1002/adma.201402996

También podría gustarte