Subscriber access provided by UNIV OF WESTERN ONTARIO
Review
Polymer-Based Nanomaterials for Photothermal Therapy: From Light-Responsive to Multifunctional Nanoplatforms for Synergistically Combined Technologies Filippo Pierini, Pawe# Nakielski, Olga Urbanek, Sylwia Paw#owska, Massimiliano Lanzi, Luciano De Sio, and Tomasz Aleksander Kowalewski Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.8b01138 • Publication Date (Web): 19 Sep 2018 Downloaded from http://pubs.acs.org on September 21, 2018
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1
Polymer-Based
Nanomaterials
2
Therapy: From Light-Responsive to Multifunctional
3
Nanoplatforms
4
Technologies
for
for
Synergistically
Photothermal
Combined
5 6
Filippo Pierini,*† Paweł Nakielski,† Olga Urbanek,§ Sylwia Pawłowska, † Massimiliano
7
Lanzi,‡ Luciano De Sio || and Tomasz Aleksander Kowalewski†
8
9
†
Department of Biosystems and Soft Matter, Institute of Fundamental Technological
10
Research, Polish Academy of Sciences, Warsaw 02-106, Poland
11
§
12
Polish Academy of Sciences, Warsaw 02-106, Poland
13
‡
14
Bologna, 40136 Bologna, Italy
15
||
16
Corso della Repubblica 79, 04100 Latina, Italy
Laboratory of Polymers and Biomaterials, Institute of Fundamental Technological Research,
Department of Industrial Chemistry “Toso Montanari”, Alma Mater Studiorum-University of
Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome,
17
18
1 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
19
Abstract. Materials for the treatment of cancer have been studied comprehensively over the
20
past few decades. Among the various kinds of biomaterials, polymer-based nanomaterials
21
represent one of the most interesting research directions in nanomedicine, because their
22
controlled synthesis and tailored designs make it possible to obtain nanostructures with
23
biomimetic features and outstanding biocompatibility. Understanding the chemical and
24
physical mechanisms behind the cascading stimuli-responsiveness of “smart” polymers is
25
fundamental for the design of multifunctional nanomaterials to be used as photothermal
26
agents for targeted polytherapy. In this review, we offer an in-depth overview of the recent
27
advances in polymer nanomaterials for photothermal therapy, describing the features of three
28
different types of polymer-based nanomaterials. In each case, we systematically show the
29
relevant benefits, highlighting the strategies for developing light-controlled multifunctional
30
nanoplatforms that are responsive in a cascade manner and addressing the open issues by
31
means of an inclusive state-of-the-art review. Moreover, we face further challenges and
32
provide new perspectives for future strategies for developing novel polymeric nanomaterials
33
for photothermally assisted therapies.
34 35 36
Keywords: polymer nanostructures; multifunctional nanomaterials, cascading stimuli-
37
responsiveness; light-to-heat conversion; photothermal therapy; targeted polytherapy for
38
cancer.
39 40 41
2 ACS Paragon Plus Environment
Page 2 of 84
Page 3 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
42
1. Introduction
43
Cancer is a class of more than one hundred diseases involving an out-of-control growth of
44
abnormal cells in the human body.1 The number of people suffering from cancer is
45
dramatically high. The World Health Organization (WHO) estimates that cancer is the second
46
leading cause of death worldwide, and that it was responsible for 8.8 million deaths in 2015.2
47
Moreover, it accounted for approximately 16% of all the world’s deaths in 2015 and it is
48
expected to continue rising. The estimated annual worldwide cost of cancer to society, 1.16
49
trillion US dollars, highlights the enormous impact that it has on global socioeconomic
50
conditions.3
51
Cancer has been recognized as a serious disease since ancient times. Hippocrates, the father of
52
medicine, classified and studied several kinds of cancer more than 2,000 years ago. During his
53
studies, he investigated different types of treatment, including the cauterization of surface
54
tumors by using a hot metal, concluding in the end that “if a tumor cannot be cut, it should be
55
burned. If it cannot be burned, then it is incurable”.4
56
The theory that cancer may be cured by artificially controlling body temperature has been
57
evaluated scrupulously over the last few decades. Many studies underlined the fact that
58
normal and cancer cells have different responses to heat, and that temperatures over 42.5°C
59
are actually cytotoxic for tumor cells.5 This concept paved the way to a new treatment called
60
hyperthermia therapy.6 Conventional hyperthermia systems are designed to heat tissue to
61
around 42.5-44.0°C in order to stimulate the immune system response for the non-specific
62
immunotherapy of cancers and to induce the regression or complete disappearance of the
63
cancer by direct cell damage.7 Unfortunately, in clinical conditions the use of hyperthermia as
64
a stand-alone treatment of cancer has been proven ineffective.8 High temperatures can
65
eliminate a large number of tumor cells, but it is difficult to heat the local tumor region
66
without causing damage to the surrounding tissues. Hyperthermia exhibits several technical 3 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
67
problems mainly related to the heating of relevant portions of the human body and to the
68
serious side effects caused to patients.9 Lastly, this technique is only applied as an additional
69
therapy, in combination with conventional cancer treatments (e.g. chemotherapy and
70
radiotherapy).8
71
The development of effective tumor treatments is one of the greatest challenges in healthcare
72
today. Therefore, huge investments have been made in research with a view to developing
73
new treatments or improving the already-existing ones. Scrupulous attention has recently been
74
focused on the application of heat in cancer treatments, with the possibility to “burn” a tumor
75
at the cellular level, thus reducing the main drawbacks of conventional hyperthermia. The key
76
to the efficient and successful treatment of a tumor using heat is the development of a non-
77
invasive therapy that leads to the local treatment of cancer cells only.10
78
Photothermal therapy (PTT) is a newly developed and promising therapeutic strategy which is
79
considered the natural evolution of conventional hyperthermia therapy, because it uses agents
80
capable of physically reaching specific cancer cells and generating heat because of the
81
interaction with light.11 PTT involves the use of nanometer sized materials that show high
82
absorption in the near-infrared (NIR) region.12 This biological tissue-transparent radiation can
83
penetrate living tissues without causing damage. The light absorbed by these nanomaterials is
84
rapidly converted into heat, causing tumor cell apoptosis (Figure 1a) and avoiding the typical
85
drawbacks of hyperthermia.10
86
The development of future medical technologies capable of enhancing therapeutic efficiency
87
and reducing their side effects will mainly be the result of the enormous scientific effort put
88
into material science, which has led to the rapid development of nanotechnology.13 Today, the
89
rapid expansion of this field has led to a generation of biomaterials with fascinating properties
90
and activities.14 Unfortunately, the first generations of biomaterials already applied were
91
designed without fully taking into consideration the fast and significant changes that occur in 4 ACS Paragon Plus Environment
Page 4 of 84
Page 5 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
92
biological tissues. The possibility to use external stimuli to trigger changes in biomaterial
93
properties affords a high level of control over the activities of implanted material over time,
94
which is necessary for developing a new generation of biomedical materials.15 One of the
95
main goals of nanomaterial sciences for biomedical application is that of developing
96
biocompatible materials in which changes in properties and activities are triggered through
97
artificial stimuli, such as light irradiation,16 magnetic,17 pH,18 thermal,19 and electrical
98
stimulations.20
99
The complex functions of living systems are mainly driven by regulation systems which
100
provide feedback to stabilize such extremely dynamic and non-equilibrium systems.21 Nature
101
uses different kinds of approaches to perform life processes precisely, based on response
102
cascades.22 Nature has been a source of inspiration for scientists in the development of new
103
technologies and materials, since it offers a wide range of multifunctional materials capable of
104
responding to stimuli in a controlled way.23 In the past decades, biomimetic approaches have
105
been used by scientists to develop stimuli-responsive materials which try to mimic nature.24
106
Sensitive materials which respond to small environmental condition changes with dramatic
107
material property variations are known as “stimuli-responsive” or “smart” or “intelligent”
108
materials. Smart nanomaterials can be classified according to the stimuli they are sensitive to
109
(e.g. temperature, pH, ionic strength, light, electric or magnetic field).25 Moreover, some of
110
these materials can respond to a combination of different kinds of stimuli.26 Stimuli-
111
responsive materials became essential in the area of biomedical applications in order to
112
develop brand new diagnostic and therapeutic techniques, and especially nanomaterials, since
113
nanotechnology shows remarkable potential in this field.27
114
Nanomedicine is an extremely dynamic field and PTT, due to its non-invasive character for
115
the selective treatment of tumors, is one of its most studied branches. Several scientific
116
articles published by different research groups have proven the possibility to synthesize 5 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
117
nanomaterials with interesting optical properties in the near-infrared region, such as the fast
118
and efficient light-to-heat conversion that can be delivered to specific tissues. The unique
119
optical properties of nanomaterials confirm the potential for the creation of new functional
120
materials for photothermal cancer therapy.28 Recently, several kinds of nanomaterials that are
121
good absorbers of NIR radiation have been developed. The first and most widely studied
122
group of materials is composed of noble metal nanomaterials (e.g. gold and silver
123
nanoparticles).29 The surface plasmon resonance – a phenomenon which leads to the
124
collective excitation of electrons in these metal nanostructures – has been widely examined to
125
strongly enhance the material’s optical response, such as the NIR radiation absorption,
126
scattering, and conversion.30 Despite the huge efforts of the scientific community to improve
127
metal nanomaterials for PTT, the accumulation of such metallic nanoparticles in the body and
128
their potential health risk could be a problem that prevents the large-scale application of these
129
nanomaterials.31 For this reason a number of research studies have been focused on the use of
130
different structures, such as carbon nanomaterials. Carbon nanotubes, as well as graphene and
131
its derivatives, have outstanding optical properties and have been indicated as potential
132
therapeutic agents for PTT applications.32 However, the practical application of one- and two-
133
dimensional carbon nanomaterials for nanomedicine is still open to doubt, since several
134
studies have demonstrated their harmful effects on body tissues and cells due to their intrinsic
135
toxicity and low biocompatibility.33
136
In the light of potential advantages, the possibility to develop organic nanomaterials as PTT
137
agents has recently attracted the attention of the scientific community (Figure 1b). Polymeric
138
nanostructured materials have been widely applied in nanomedicine for various purposes such
139
as tissue engineering,34 drug delivery,35 biosensors,36 antibacterial materials,37 and
140
biointerfaces.38 Several kinds of polymer nanomaterials possess the main requirements
141
necessary to be considered as potential PTT agents: suitable size and shape, dispersibility in 6 ACS Paragon Plus Environment
Page 6 of 84
Page 7 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
142
an aqueous medium, light-to-heat conversion of NIR radiation, photostability, low
143
cytotoxicity and accumulation in living tissues.39,
144
application are not only more biodegradable and remain in the body for shorter periods of
145
time after systemic administration than inorganic nanoparticles,41 but they also offer indirect
146
advantages because of their synthesis and the possibility to apply further forms of treatment
147
with a view to developing more advanced biomaterials.
148
“Green chemistry” is an area of chemistry focused on the development of sustainable
149
technologies for the production of materials by chemical strategies. “Green Polymer
150
chemistry”, which is at the forefront of this innovative strand of work, has led to the
151
development of a few encouraging viable synthetic strategies for suitable polymer synthesis.42
152
Moreover, unlike inorganic nanomaterials, polymers can be easily processed using post-
153
synthesis treatments for the preparation of nanomaterials with well-defined structures.
154
Advanced techniques like electrospinning
155
with great success to fabricate biomaterials for medical applications. Most importantly,
156
tailored polymer-based nanomaterials could be designed to produce a cascade response to
157
light stimulations.45 Using this nature-inspired mechanism, light irradiation activates the
158
material response (e.g. heat generation), which acts as a “mediator” to trigger structural
159
polymer transformations (e.g. expansion/contraction, chain mobility, crystallinity, or material
160
rupture) and in turn gives novel material functionalities such as on-off switching of drug/gene
161
delivery or signals for imaging purposes. This unique and fascinating property makes it
162
possible to developed multifunctional materials which are able to combine photothermal
163
therapy with other targeted cancer diagnostic and therapeutic treatments.46
43
40
Polymer-based nanomaterials for PTT
and 3D-printing
164
7 ACS Paragon Plus Environment
44
have already been applied
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
165 166
Figure 1. (a) Scheme illustrating the biochemical mechanism resulting from nanomaterial-
167
mediated photothermal therapy. Reprinted with permission from ref. 10. Copyright 2015
168
American Chemical Society. (b) The number of published articles on polymer-based
169
nanomaterials for photothermal therapy during the period 2006–2017, obtained from Web of
170
Science database.
171 172
The main aim of this review is to summarize the potential of polymer-based nanomaterials as
173
photothermal cancer therapeutic agents for targeted polytherapy. We summarize the recent
174
results in designing, fabricating, and testing different types of nanostructures which show a
175
potential for developing multifunctional nanoplatforms thanks to their cascading
176
responsiveness to light stimuli. Finally, we highlight the key points for the development of
177
more effective nanomaterials and the emerging perspectives and challenges for future
178
research from the viewpoint of material scientists.
179 180 181 182
8 ACS Paragon Plus Environment
Page 8 of 84
Page 9 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
183
2. Conjugated Polymer Nanomaterials
184
Conjugated polymers (CPs) are an important class of macromolecules having the π-electrons
185
system delocalized over the whole polymeric main chain. Since the discovery of the doping of
186
polyacetylene, many other new polymers, with a high structural versatility and stability in the
187
neutral and the doped states, have been successfully synthesized and applied in different fields
188
such as batteries,47 organic electronics,48 and solar cells.49 Moreover, these polymers have
189
recently attracted great interest due to their biomedical applications.50
190
Photothermal therapy exploits photo-absorbing agents which are able to efficiently convert
191
the absorbed light into heat, in order to induce a local hyperthermia that results in irreversible
192
damage for cancer cells. In a similar way, photodynamic therapy (PDT) uses photosensitizers
193
to transfer coherent light energy to oxygen molecules, thus generating cytotoxic reactive
194
oxygen species (ROS). While photosensitizers for PDT can also be activated using visible
195
light, an important prerequisite for PTT is the use of CPs that absorb in the NIR region
196
(650-1700 nm), since blood and tissues absorb in the UV-Vis spectral range. Recently, CPs
197
have been successfully used for PTT and PDT in cancer thanks to their highly electronic-
198
delocalized structures, which are particularly prone to absorbing light to be converted to heat,
199
fluorescence, and other energies. Moreover, some CPs nanosystems show photoacoustic (PA)
200
effects. Basically, they are able to absorb light, creating a thermally-induced pressure increase
201
which generates ultrasonic waves (phonons). Phonons can be received by acoustic detectors to
202
form high-resolution images of deep tumors.
203
A new important class of CPs is D-A copolymers, which have a combination of electron-rich
204
(ED) and electron-deficient (EA) moieties in their conjugated backbone. They usually show
205
very low band-gap (π-π*) energy and long wavelength absorptions. Photons in the NIR-II
206
region (950-1700 nm) provide more efficient tissue penetration ability than NIR-I photons
9 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
207
(650-950 nm), when considering absorption and scattering effects in tissues. Moreover, the
208
structural versatility of CPs makes the insertion of specific chromophores, photosensitizers, or
209
other functional groups of biological interest (in the polymer main chain or in side chains)
210
particularly simple. CPs are usually hydrophobic materials, but they can be effectively coated
211
by hydrophylic polymers in order to achieve water-dispersable nanoparticles (NPs) for
212
biomedical applications. CP NPs can also be loaded with a variety of photothermal agents,
213
metallic NPs, and anticancer drugs by conventional nanoencapsulation strategies. Herein, we
214
describe some examples of photothermal (PT) and photodynamic (PD) therapeutic CP
215
nanosystems, together with recent developments in combinational drug-assisted therapy and a
216
summary of currently-available studies involving the application of these multifunctional
217
nanoplatforms is presented in Table 1.
218 219
2.1 Polyaniline
220
Polyaniline (PANI) can easily transit from emeraldine base (EB) state to emeraldine salt (ES)
221
state in the presence of strong acids with a marked red-shift of its absorption maximum
222
wavelength toward the NIR region. Molina et al. synthesized a thermoresponsive
223
nanocomposite incorporating in-situ polymerized PANI.51 The dendritic nanogel, based on
224
poly(N-isopropylacrylamide) (PNIPAM) crosslinked with 33% wt. of dendritic polyglycerol
225
(dPG), was swollen in a solution of aniline and polymerized using (NH4)2S2O8. The final
226
interpenetrated nanogel showed an excellent compatibility in physiological environments, and
227
in vivo studies showed that mice tolerated an accumulated dose of 500 mg kg-1, which is able
228
to inhibit A2780 tumor transplant growth by 55.3% after exposition to NIR laser light
229
(distance: 5 cm, 5 min, 0.5 W maximum power). A simple method for preparing hybrid PANI
230
NPs has been reported by Wang et al.52 EB PANI was doped with n-dodecylbenzenesulfonic
231
acid (DBSA), yielding ES PANI, which was dissolved in CHCl3 and added to 10 ACS Paragon Plus Environment
Page 10 of 84
Page 11 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
232
1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC). After solvent evaporation, the thin
233
film of Lipid-PANI NPs was conjugated with folic acid (FA) as the targeting ligand to
234
enhance its tumor-targeting ability. FA-Lipid-PANI NPs showed high biocompatibility and
235
stability in physiological environments and – owing to the strong absorbance in the NIR
236
region (λmax=810 nm), which is ascribable to the presence of polarons in the conjugated PANI
237
backbone – showed a relatively high photothermal conversion efficiency (η=25.6%) under
238
laser irradiation at 808 nm. The in vitro cytotoxicity of NPs toward HeLa (human cervical
239
cancer cells) reached nearly 70% at a NPs concentration of 100 µg ml-1 under 808 nm
240
irradiation. Moreover, the local tumor temperature of HeLa tumor-bearing BALB/c mouse
241
treated with tail injections of FA-Lipid-PANI NPs rapidly increased up to 50°C upon
242
irradiation (@ 808 nm, 2 W cm-2, 5 min) and the tumor was completely eliminated after 10
243
days.
244 245
2.2 Polypyrrole
246
Polypyrrole (PPy) is one of the most studied ICPs for PTT applications thanks to its strong
247
absorbance in the NIR region. Recently, Bharathiraja et al. have developed PPy NPs using
248
astaxanthin (Astx) conjugated bovine serum albumin (BSA) as a coating agent
249
(PPy@BSA-Astx).53 They used Astx, a natural keto-carotenoid pigment, as a photosensitizer
250
to generate ROS (reactive oxygen species) under 808 nm laser irradiation. At a power density
251
of 0.3 W cm-2 for 15 min they observed a significant reduction of MDA-MB-231 cell
252
viability, depending on the nanoparticle concentration, which is ascribable to the
253
photodynamic toxic effect on breast cancer. Moreover, PPy@BSA-Astx NPs in water solution
254
at 50 µg mL-1 concentration showed a high photothermal efficiency imputable to PPy,
255
enabling the possibility to reach 57°C in 5 min at 1 W cm-2 power density. NPs were also
11 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
256
screened for ultrasound signal generation under 808 nm laser irradiation, revealing their
257
ability to generate PA signals even at low concentrations (10 µg mL-1). Since the NIR II
258
window offers a high efficiency in tissue penetration owing to the reduced photon scattering
259
and lower tissue background, Wang et al. recently prepared ultrathin PPy nanosheets with
260
improved absorption at 1064 nm
261
second NIR window (Figure 2).54 PPy is a low-cost, biocompatible, photostable conjugated
262
polymer which can be easily doped up to high levels with a controlled doping process by
263
means of Fe(III) inorganic salts. A layered iron oxychloride template was used to obtain
264
ultrathin PPy nanosheets in which the CP was in its doped, low-bandgap, high-wavelength
265
absorption bipolaron state. The as-synthesized nanosheets were coated with 1,2-distearoyl-sn-
266
glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000),
267
obtaining highly stable physiological dispersions for biomedical applications. PPy nanosheets
268
exhibited high in vitro and in vivo photothermal ablation ability toward cancer cells in the
269
NIR II window. Their photothermal conversion efficiency was 55.6% at 808 nm and even
270
higher (64.6%) at 1064 nm; in vitro studies revealed that 80% of MDA-MB-231 human breast
271
adenocarcinoma cells were killed when exposed to a 1064 nm laser beam (1.0 W cm-2, 10
272
min). Moreover, the use of PPy nanosheets permitted the effective PT ablation of tumors in
273
nude mice at depths of 8 and 6 mm, by means of 1064 and 808 nm laser sources respectively,
274
suggesting promising applications of these nanomaterials for effective deep-tissue PTT.
(ε=27.8 L g-1 cm-1) to be used as PT agents in the
12 ACS Paragon Plus Environment
Page 12 of 84
Page 13 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
275 276
Figure 2. (a) Schematic illustration of the preparation of PPy nanosheets. (b) Fluorescent
277
images at different time points post intravenous injection of PPy nanosheets. (c) Infrared
278
thermographic images of tumor-bearing mice after intravenous injection of PBS and PPy
279
nanosheets after irradiation by a 1064 nm laser. Reprinted with permission from ref. 54.
280
Copyright 2018 American Chemical Society.
281 282
2.3 Polythiophene
283
Polythiophene (PTh) derivatives usually exhibit excellent photostability, light-harvesting
284
ability, easy synthesis, and facile functionalization with different substituents. Ren et al.
285
successfully synthesized a new organic compound by covalently linking 2-N,N’-bis(2-
286
(ethyl)hexyl)perylene-3,4,9,10-tetra-carboxylic acid bis-imide to a thienylvinylene oligomer,
287
obtaining a conjugated copolymer (C3) with excellent photothermal properties, thanks to the
288
presence of benzene rings and conjugated double bonds (Figure 3a).55 C3 was coprecipitated 13 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
289
with PEG-PCL and indocyanine green (ICG) in order to obtain encapsulated multifunctional
290
nanospheres with an average diameter of 50 nm. When PEG-PCL-C3-ICG NPs distilled water
291
solution was exposed to 808 nm laser irradiation for 10 min, the temperature increased from
292
25°C up to 47°C, confirming the strong PT effect. Moreover, multifunctional encapsulated
293
NPs exhibited a low cytotoxicity and an efficient ROS production in vitro for PDT treatment
294
of oral squamous cell carcinoma (OSCC). Additionally, HSC tumor-bearing mice injected
295
with NPs and exposed to laser radiation (@808 nm, 2 W cm-2, 8 min) showed a remarkable
296
tumor volume reduction. The persistence of conjugated polymer nanoparticles (CPNs) could
297
be a threat to human body. In fact, an important challenge for the use of CPNs for in vivo
298
theranostic is to prevent accumulation of nanoparticles in the body. Recently, Lyu et al.
299
developed a biodegradable conjugated polymer which can be gradually broken down into
300
small fragments in presence of the oxidative species normally abundant in the biological
301
environment.56
302
tetrahydropyrrolo[3,4-c]pyrrole-1,4-diyl)-dithiophene]-5,5′-diyl-alt-vinylene} (DPPV) (Figure
303
3b), the presence of vinylene bonds in the main chain not only enhances its biodegradabilty
304
and biocompatibility, but also increases the photothermal conversion efficiency and the mass
305
absorption coefficient, for a superior antitumor efficacy. Moreover, DPPV showed an intense
306
PA activity, for a more effective imaging capability. The PTT conversion efficiency of
307
DPPV/PLGA-PEG NPs reached a notably high value of 71% (@ 808 nm, 0.3 W cm-2),
308
among the best values recorded for photothermal theranostic agents. Antitumor effect of
309
DPPV PEGylated NPs was evaluated by measuring the volumes of tumors induced by
310
injecting 4T1 mammary carcinoma cells in living mice, resulting that the tumor growth was
311
effectively inhibited during the whole tested period. Finally, the ex-vivo histological data
312
evidenced that no significant histopathological lesions or abnormalities were observed for the
In
the
synthesized
poly{2,2′-[(2,5-bis(2-hexyldecyl)-3,6-dioxo-2,3,5,6-
14 ACS Paragon Plus Environment
Page 14 of 84
Page 15 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
313
main vital organs of treated mice, owing to the good biodegradability and organically benign
314
composition of the employed CPNs.
315
316 317
Figure 3. (a) Synthetic route of C3. Reprinted with permission from ref. 55. Copyright 2017
318
American Chemical Society. (b) Synthesis of DPPV ((i) Pd2(dba)3 and tri(o-tolyl)phosphine,
319
toluene, 100°C, 24 h) and schematic illustration of DPPV/PLGA-PEG NPs. Reprinted with
320
permission from ref. 56. Copyright 2018 American Chemical Society.
321 322
2.4 CPs containing organic light-absorbing dyes
323
Porphyrins are strong light-absorbers, showing an absorption band around 400 nm (Soret
324
band) and two Q-bands around 510 and 540 nm. Recently, Wang et al. reported on the
325
synthesis of polymeric NPs loading ICG, Pt(II)-meso-tetra(pentafluorophenyl)porphyrine
326
(PtTFPP) and poly(9,9-di-n-octylfluorenyl-2,7-diyl) (PFO) (Figure 4).57 The simultaneous
327
presence of a photo-sensitizer (PtTFPP), an organic semiconducting polymer acting as two-
328
photon antenna (PFO), and a NIR-absorbing photothermal dye (ICG) in the same nanoparticle
329
led to a strong photothermal performance under 808 nm laser irradiation and a high 1O2
15 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 84
330
generation efficiency under NIR light exposition, thus making the assembled multifunctional
331
therapeutic nanoplatform particularly promising for cancer treatment.
332 333
Figure 4. Schematic illustration of multifunctional ICG-PtTFPP NPs. Reproduced from ref.
334
57. Republished with permission of Royal Society of Chemistry, Copyright 2017; permission
335
conveyed through Copyright Clearance Center, Inc.
336 337
2.5 D-A conjugated polymers
338
D-A conjugated polymers are very useful materials for photothermal cancer therapy. Indeed,
339
their HOMO-LUMO gap can be easily tuned by the choice of the suitable electron
340
donor/electron acceptor moieties. Cao et al. recently designed a thieno-isoindigo derivative-
341
based
342
bis(5-oxothieno[3,2-b]pyrrole-6-ylidene)benzodifurandione
343
3,3’-didodecyl-2,2’-bithiophene (BT) units acted as EA and ED, respectively (Figure 5a).58
344
The polymer, with a very low bandgap (1.24 eV) and a λmax at 1107 nm, was formulated into
345
nanoparticles using an amphiphilic diblock copolymer to enhance its solubility in aqueous
346
environments. The obtained NPs showed a spherical morphology with a diameter of about 45
semiconducting
polymer,
PBTPBF-BT,
16 ACS Paragon Plus Environment
in (BTPBF)
which
the
and
the
Page 17 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
347
nm and a remarkable mass extinction coefficient of 89.3 mL cm-1 mg-1 at 1064 nm, in the NIR
348
II region. NPs have been evaluated as PT agents by exposing MDA-MB-231 cells to a 1064
349
nm laser at a power density of 0.42 W cm-2. After 10 minutes, more than 50% of the cancer
350
cells were killed using a NPs concentration of 3.75 µg mL-1. In vivo activity was tested on
351
MDA-MB-231 tumor-bearing mice which were injected with a PBTPBF-BT NPs dose of 30
352
µg per mouse and exposed to 1064 nm laser light for 10 min at 0.42 W cm-2. The tumor
353
growth was completely inhibited and only black scars were observed at the original tumor
354
sites at 14 days post-treatment. Jiang et al. synthesized a conjugated copolymer with
355
alternated cyclopentadithiophene (ED) and benzothiadiazole (EA) units. Hydrophilic PEG
356
chains were grafted onto hydrophobic CP backbone leading to water-soluble NPs.59 The
357
presence of CP in NPs gave them some important properties: an intense PT activity, a strong
358
NIR fluorescence, and the ability to interact with the aromatic anticancer Doxorubicin (DOX)
359
through hydrophobic π-π interactions. Yuan et al. also described the preparation of a
360
multifunctional nanoplatform by combining DOX with a PT conjugated polymer based on
361
thiadiazoloquinoxaline EA units alternated with functionalized fluorene ED units.60 The use
362
of a NIR laser-responsive amphiphilic brush copolymer as the encapsulation matrix allowed
363
for an efficient phototriggered drug release. The obtained NPs were also surface-modified
364
with cyclic arginine-glycine-aspartic acid (cRGD) tripeptide in order to increase their
365
accumulation within cancer cells. Feng et al. designed an organic NP-based theranostic
366
platform using two D/A copolymers: poly[9,9-bis(2-(2-(2-methoxyethoxy)ethoxy)-ethyl)
367
fluorenyldivinylene]-alt-4,7-(2,1,3-benzothiadiazole) (PFVBT), with bright red fluorescence
368
and high ROS production, and poly[(4,4,9,9-tetrakis(4-(octyloxy)phenyl)-4,9-dihydros-
369
indacenol-dithiophene-2,7-diyl)-alt-co-4,9-bis(thiophen-2-yl)-6,7-bis(4-(hexyloxy)phenyl)-
370
thiadiazolo-quinoxaline] (PIDTTTQ), with a broad absorption in the NIR region.61 The two
371
copolymers were coencapsulated to form CP NPs using a PEG matrix, and an anti-HER2
17 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
372
affibody was attached to the nanoparticle surface through surface conjugation. The obtained
373
multifunctional theranostic NPs showed high fluorescence (η=23%) and light-to-heat
374
conversion efficiency (47.6%), and an excellent targeting ability toward HER2 overexpressed
375
SKBR-3 breast cancer cells. A D-A porphyrin-containing conjugated polymer showed the
376
absorption peak at 799 nm, good biocompatibility and photostability, and a very high PT
377
conversion efficiency (63.8%).62 The in vitro cancer cell ablation activity of its NPs was
378
satisfying, since at the concentration of 5 mg L-1, almost 80% of MDA-MB-231 human breast
379
cancer cells were killed under laser irradiation at 0.75 W cm-2 for 10 min. Conjugated
380
oligomers often exhibit a better-defined structure and a higher degree of purity and synthetic
381
reproducibility than their conjugated polymer analogs. Cai et al. recently developed novel D-
382
A conjugated oligomer-based NPs with high PT activity and biocompatibility and an excellent
383
contrast when used for PA imaging of sentinel lymph nodes.63 An efficient NIR II PT
384
conversion was obtained using the new D-A conjugated polymer TBDOPV-DT.64 It contained
385
a thiophene-fused benzodifurandione-based oligo(p-phenylenevinylene) (TBDOPV) as EA
386
unit and 2,2’-bithiophene (DT) as ED moiety (Figure 5b), and showed an absorption peak at a
387
very long wavelength (1093 nm), a high biocompatibility, and a highly efficient photothermal
388
conversion through 0.2 cm of pig skin tissue (33% of transmittance).
389
390
18 ACS Paragon Plus Environment
Page 18 of 84
Page 19 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
391
Figure 5. (a) Schematic illustration of PBTPBF-BT NPs. Reprinted from ref. 58. Copyright
392
2017, with permission from Elsevier. (b) Chemical structure of TBDOPV-DT polymer.
393
Reprinted with permission from ref. 64. Copyright 2017 American Chemical Society.
394
Huang and coworkers reported on the synthesis of a theranostic agent based on
395
benzo[1,2-c;4,5-c’]bis[1,2,5]thiadiazole-4,7-bis(9,9-dioctyl-9H-fluoren-2-yl)thiophene
396
(BBT-2FT) to take advantage of the high EA characteristics and strong light absorption of
397
benzo-bis-thiadiazole (BBT) derivatives.65 The conjugated molecule contained fluorene
398
moieties as ED groups and a BBT unit as electron-acceptor. The presence of hypervalent
399
sulfur atoms in quinoidal structures in the conjugated backbone ensured high NIR absorption
400
in the NIR II therapeutic optical window. By coprecipitating the conjugated molecule in the
401
presence of the block copolymer PEG-b-PCL, they obtained BBT-2FT NPs, which showed
402
high PA signal intensity and PT conversion efficiency (η=40%). When CPs are encapsulated
403
into NPs and suspended in aqueous media, their aggregation can reduce the overall
404
fluorescence and the ROS generation ability. However, some types of weakly emissive NPs
405
show the opposite phenomenon, being induced to emit by aggregate formations. Another D-A
406
conjugated polymer architecture for PTT was proposed by Li et al.66 PBIBDF-BT copolymer
407
showed in its structure the EA isoindigo derivative bis(2-oxoindolin-3-ylidene)-benzodifuran-
408
dione (BIBDF) and the ED unit bithiophene (BT). NPs were obtained by emulsifying the
409
conjugated polymer with the amphiphilic copolymer mPEG-b-PHEP and showed a high
410
antitumor efficiency toward
411
0.5 W cm-2, 10 min). A higher activity was reached when PBIBDF-BT NPs were coloaded
412
with doxorubicin, since NIR irradiation was capable of efficiently triggering DOX release
413
from PBIBDF-BT@NPPPE/DOX nanoparticles. Combining D-A conjugated polymer with
414
wide-absorption organic dyes can be an efficient way to improve the light-absorbing ability of
415
the polymer to further enhance its theranostic properties, especially for PT applications.
MDA-MB-231 tumors in mice by irradiation (808nm,
19 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
416
Recently, for the first time, Zhang et al. introduced a light-harvesting unit on a D-A
417
polymer.67 The semiconducting polymer was obtained by the copolymerization of 5,6-
418
difluoro-4,7-bis[5-(trimethylstannyl) thiophen-2-yl]benzo-2,1,3-thiadiazole as an EA and 5,5′-
419
dibromo-4,4′-bis(2-octyldodecyl)-2,2′-bithiophene as an ED in the presence of a porphyrin-
420
pyrene pendant as a side-branch unit. The obtained copolymer was nanoprecipitated with
421
HOOC-PEG-COOH and PMHC18-mPEG copolymers to increase the water dispersibility and
422
size stability of the resulting biocompatible
423
semiconducting polymer nanoparticles NPs. As expected, the spherical-shape NPs showed a
424
very high photothermal conversion (η=62.3%) and a strong fluorescence for PA imaging-
425
guided PTT. Mice injected with A549 human lung cancer cells achieved 100% tumor
426
elimination using a dose of 15 mg kg-1 of NPs under laser irradiation (630 nm, 0.8 W cm-2, 10
427
min). Although many polymeric photothermal agents have shown great potential for cancer
428
treatment, they are often assembled in unstable nanostructures which can degrade in short
429
times. Chang et al. recently synthesized a new D-A copolymer: 4,8-bis[5-(2-
430
ethylhexyl)thiophen-2-yl]-2,6-bis(trimethylstannyl)benzo[1,2-b:4,5-b′]dithiophene-6,6′-
431
dibromo-N,N′-(2-ethylhexyl)isoindigo (BDT-IID) with a strong absorption in the 650-700 nm
432
range.68 Polymer dots, with spherical morphology and an average diameter of 18 nm, were
433
obtained by coprecipitating BDT-IID and poly(styrene-co-maleic anhydride) (PSMA).
434
Polymer dots (Pdots) aqueous dispersions were very stable and no aggregation was observed
435
even after 30 days while they showed an enhanced photostability. NPs dispersions in water at
436
a concentration of 100 µg mL-1 were irradiated using a laser source (@ 660 nm, 200 mW cm-
437
2
438
still maintained the ability to increase the temperature to the same level, denoting an excellent
439
photostability of the employed chromophoric system. Moreover, since the measured
440
photothermal conversion efficiency was particularly high (45%), the prepared nanomaterials
electron donor–acceptor conjugated
) reaching a temperature of 53.2°C after 600 s. Even after seven irradiation cycles, the Pdots
20 ACS Paragon Plus Environment
Page 20 of 84
Page 21 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
441
were also examined as PT agents for tumor therapy. Animal tests were carried out on MCF-7
442
tumor-bearing mice injected with BDT-IID Pdots and subjected to irradiation under the
443
previously described conditions. The results showed a marked tumor inhibition when
444
compared to the control group mice. More importantly, no appreciable signs of organ
445
damages or inflammatory lesions could be observed on major organs as a consequence of the
446
PTT treatment. All the aforementioned results demonstrated that the BDT-IID Pdots are
447
particularly promising for nanotheranostic applications. A few novel nanoplatforms based on
448
Pdots for tumor phototherapy has been developed by the Wu research group.69,70 Recently,
449
Chen et al. designed a conjugated polymer-based nanocarrier for enhanced anticancer PTT by
450
integrating a diketopyrrolopyrrole-dithiophene (DPP-DT) derivative with an amphiphylic
451
polymer (polystyrene-grafted ethylene oxide functionalized with carboxyl groups) used as the
452
encapsulation matrix.69 DPP-DT NPs (Pdots) were prepared starting from semiconducting
453
polymer solutions at different concentrations, observing that NPs with larger sizes were
454
recovered when the concentration of the solution was increased. Interestingly, the wavelength
455
of the absorption peak of Pdots was strongly dependent on their size and also on the
456
molecular weight of the semiconducting polymer. This very versatile system was capable of
457
tuning its optical absorption by simply acting on particle morphology and on the mean
458
conjugation length of the conjugated polymer. PEGylated polymer dots showed high PT
459
conversion efficiency (>50%), photostability and therapeutic performance. Mice bearing H22
460
or 4T1 tumors were injected with coated Pdots (1.0 mg kg-1) and subsequently exposed to
461
laser irradiation
462
inhibited, leaving only black scars at the original tumor sites which faded in about 10 days.
463
Fluorescence imaging technique, offering high temporal resolution, safety and sensitivity at a
464
low cost, has been extensively investigated for the purpose of diagnostics and therapeutic of
465
diseases. Conjugated Pdots have been widely accepted as a promising class of fluorophores
(@ 808 nm, 0.5 W cm-2, 5 min) and tumor growth was strongly
21 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
466
due to their intense brightness and nontoxic features. Moreover, these particles could emit in
467
the NIR region acting as local sources of activation for PTT agents. Wu et al. synthesized
468
polymer-blended Pdots of a green-light-harvesting polymer (poly[(9,9-dioctylflurenyl-2,7-
469
diyl)-co-(1,4-benzo-{2,1’,3}-thiadiazole)] (PFBT)) as the ED unit and a deep-red emitting
470
polymer
471
95:5 (PF-DBT5)) as the EA unit.71 Pdots showed a high absorption in the visible range and
472
high quantum yield in deep-red emission. PF-DBT5, PFBT and an amphiphilic polymer
473
(PSMA) were cocondensed giving highly fluorescent polymeric blend dots (PBdots) which
474
exhibited a quantum yield of 0.56, the highest value reported so far for polymer dots. PBdots
475
were then covalently conjugated to a peptide ligand CTX, a tumor-targeting agent with a
476
strong affinity for neuroectodermal tumors. PBdots-CTX were able to traverse the blood-brain
477
barrier and selectively target ND2:SmoA1 tumors in mice. They also proved to be photostable
478
during the in vivo fluorescence signal recording, but no evident fluorescence was observed in
479
mice blood 72 h after injection, indicating a good clearance of this new type of NP platform.
480
Among fluorescence imaging techniques, NIR fluorescence has been successfully employed
481
for clinical image-guided cancer surgery. In fact, NIR fluorescent probes (λ>700 nm) are
482
particular promising since they allow in vivo bioimaging with high penetration depth and
483
minimal auto-fluorescence interference. Chen et al. developed a novel semiconducting
484
polymer by incorporating a NIR emissive porphyrin unit and other monomers into one
485
polymeric backbone, using Suzuki-coupling reaction.72 The aim of the authors was to obtain a
486
new polymeric system with a large absorption coefficient in the visible region and an efficient
487
promotion of the cascade energy transfer to the NIR unit. The synthesized polymer (NIR800),
488
showing an intense emission at 800 nm with a narrow bandwidth, was further blended with
489
(poly[(9,9-dioctylflurenyl-2,7-diyl)-co-(4,7-bis(4-hexylthiophen-2-yl)-2,1,3-
490
benzothiadiazole)] (PFDHTBT) as a donor polymer, to enhance the energy-transfer via
(poly[(9,9-dioctylflurenyl-2,7-diyl)-co-(4,7-di-2-thienyl-2,1,3-benzothiadiazole)]
22 ACS Paragon Plus Environment
Page 22 of 84
Page 23 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
491
Förster mechanism. The blended Pdots showed a broad absorption range (from 500 to 600
492
nm) and an enhanced quantum yield (15%) for the narrow band emission. The PEGylated
493
Pdots evidenced a long blood-circulation time and were selectively accumulated to the lymph
494
nodes and tumors, allowing to obtain high resolution fluorescence mapping for image-guided
495
surgery.
496
Table 1. A summary of studies on the use of conjugated polymer PTT nanoplatforms in
497
cancer polytherapies.
Polymer
Wavelength of laser (nm)
Laser power density (W cm-2)
Radiation time (min)
Temp. reached (°C)
Cell line
Animal model
Ref.
PNIPAMdPG/PANI
785
-
20
42
A2780
Female nude mice
51
FA–Lipid– PANI
808
2
5
50
HeLa
Female BALB/c mice
52
PPy@BSAAstx
808
1
5
57
MDA-MB231
-
Ppy
1064
1
10
55
MDA-MB231
BALB/c nude mice
54
PEG-PCLC3-ICG
808
2
8
47
HSCs
Nude mice
55
DPPV
808
0.3
5
51
4T1 and RAW264.7
BALB/c mice
56
ICGPtTFPP
808
1
5
48
HepG2
PBTPBFBT
1064
2
5
56.4
MDA-MB231
Female BALB/c nude mice
58
DSPN5
808
1
6
60.9
4T1
BALB/c mice
59
TTTQ/DOX
800
1
10
56
MDA-MB231
-
PFVBT/PI
808
0.5
2
60
SKBR-3
Nude
23 ACS Paragon Plus Environment
-
53
57
60 61
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 24 of 84
DTTTQ
mice
PorCP
808
0.75
10
66
HeLa and MDA-MB231
Zebrafish
62
N4
808
0.8
8
64
MDA-MB231 and NIH-3T3
-
63
TBDOPVDT
1064
0.98
1
107
-
Pig skin
64
BBT-2FT
808
1.77
10
62
HeLa
-
65
PBIBDFBT
808
1
10
55
MDA-MB231
Female BALB/c nude mice
66
PPor-PEG
630
0.8
10
50
HeLa
Male nude mice
67
BDT-IID
660
0.2
10
58.8
MCF-7
Male nude mice
68
DPP-DT
808
0.5
5
62
MCF-7 and HeLa
Female nude mice
69
498 499 500
3. Polymer-Functionalized Nanoparticles
501
Polymer functionalization of nanoparticles is a widely used technique for stabilizing
502
biomaterials and preventing their aggregation. In the case of metal and semiconducting
503
nanoparticles, the inter-particle attractive forces are especially strong. Moreover, it is very
504
important to prevent the attractive interactions of such nanoparticles with the components
505
found in body tissues. This is why these nanoobjects should be fully dispersed and stable.
506
Polymer functionalization is a successful strategy for enhancing the stability of colloidal
507
dispersions.73 Several coating strategies of functionalization using polymers are well-known.
508
One of the most useful strategies is the end-grafting of polymer chains onto the nanomaterial
24 ACS Paragon Plus Environment
Page 25 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
509
surface. The formation of a polymer brush at the colloidal interface is one of the most
510
efficient methods used for colloidal dispersion stabilization.74
511
In this section we focus on the functionalization of nanoobjects by using different kinds of
512
polymers, which help to improve the chemical, physical, and biological properties of
513
nanomaterials for photothermal therapy, and endow the synthesized materials with novel and
514
unexpected functionalities.
515
3.1 PEGylation
516
Nanomaterial functionalization with poly(ethylene glycol) (PEG) is one of the most common
517
methods used to modify the surface of nanoparticles and give them stability in water,
518
biocompatibility, and other interesting features. Literature has many examples of the use of
519
this polymer to functionalize particles for biomedical applications.75 Some of them have
520
shown that during in vivo tests, nanoparticles functionalized with PEG demonstrate
521
remarkable properties such as the reduced clearance through the reticuloendothelial system of
522
the organism and the passive targeting of tumors because of the retention effect and enhanced
523
permeability. Similarly, PEGylation allows a long circulation time for NPs in the
524
bloodstream,76 while on the other hand, this technique is not useful for developing
525
multifunctional nanomaterials for polytherapy applications. These functionalized particles
526
have been investigated in numerous studies to prove their effective use as active agents for
527
photothermal therapy.
528
Li et al. proposed PEG-functionalized copper nanowires as new photothermal therapy
529
agents.77 The PEGylation chemical process consisted of adding 200 mg of PEG to 15 mg of
530
copper nanowires dispersed in 15 mL of THF. The mixture was stirred at 50°C for 4 h. The
531
functionalized nanomaterials have been characterized by higher flexibility than uncoated
532
wires and, after implantation, they are intertwined around cancer cells. Thanks to this, the
25 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
533
irradiated material transmits the generated heat to cells and effectively cause cancer ablation
534
(Figure 6a). Tests have been conducted on colon tumor-bearing mice. PEGylated
535
nanomaterials have been intratumorally injected and NIR irradiation conducted by using a
536
NIR laser (808 nm) with a power density of 1.5 W cm-2. After 6 minutes of irradiation, the
537
temperature increased rapidly above 50°C, and the suppression of tumor growth and necrosis
538
have been observed (Figure 6b).
539 540
Figure 6. (a) Scheme representing the use of PEGylated copper nanowires as a photothermal
541
agent for cancer therapy. (b) Tumor volume of CT26-bearing BALB/c mice after PEGylated
542
copper nanowires after photothermal ablation. Reprinted with permission from ref. 77.
543
Copyright 2016 American Chemical Society.
544 545
O’Neal and his co-workers used PEG-coated nanoshells for treating murine colon carcinoma
546
cells.78 To prepare PEGylated nanoshells, PEG with a terminal thiol group was added to gold-
547
silica nanoshells in deionized water. After the intravenous injection of PEGylated nanoshells
548
and their six-hour circulation in the body, mice have been illuminated with a 808 nm diode
549
laser (4 W cm-2) for 3 minutes. During the first 30 seconds, the temperature reached 50ºC.
550
After 10 days of nanoshell treatment, a complete resorption of the tumor has been observed.
551
At 90 days of post-treatment, all mice were healthy and without tumors. Similarly, gold 26 ACS Paragon Plus Environment
Page 26 of 84
Page 27 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
552
nanoshell agents functionalized with PEG have been developed by Hirsch et al.79 Yang et al.
553
have studied the possibility of using PEGylated conjugated nanographene sheets for the
554
photothermal therapy of murine breast tumors.80 The PTT agent preparation included the
555
attachment of amine-terminated branched PEG to graphene oxide sheets via amide formation.
556
Mice have been treated with 20 mg kg-1 of PEGylated nanosheets. After exposure to 2 W cm-2
557
of 808 nm NIR laser, the tumor surface temperature increased up to 50ºC. The Authors have
558
demonstrated that this treatment had completely eliminated 4T1 murine breast tumors and had
559
led to a 100% mouse survival. In comparison to non-functionalized gold nanorods, the
560
properties of PEGylated nanosheets seem to be similar in terms of administration routes
561
(intravenous), injected doses (20 mg/Kg), NIR laser densities requirements (2 W cm-2), and
562
irradiation term (5 min). Interestingly, in vivo tests also show relatively low retention in
563
reticuloendothelial systems and a highly efficient passive tumor targeting.
564
Cyanine (Cy) is an organic dye with high extinction coefficient in the NIR window which has
565
been widely adopted as a drug in PTT therapy. Lin et al. prepared a poly(heptamethine) Cy-
566
containing derivative (CyP) using a one-pot multicomponent reaction.81 CyP was
567
encapsulated into NPs using PEG-PLA copolymer obtaining high Cy-loading nanospheres
568
with an average diameter of 50 nm. CyP@PEG-PLA NPs were stable in aqueous solutions for
569
half a year, showed a PT conversion of 12% and caused irreversible tumor cellular damage in
570
mice with HeLa tumors upon 808 nm light irradiation for 30 min after injection of 150 µL of
571
a 200 µg mL-1 NPs solution.
572 573
3.2 Chitosan functionalized nanomaterials
574
Chitosan is a natural cationic polymer which is characterized by properties that are significant
575
for
biomaterials:
non-toxicity,
biodegradability,
and
27 ACS Paragon Plus Environment
biocompatibility.
Chitosan
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
576
functionalization of nanoparticles for PTT has several advantages. Indeed, chitosan coatings
577
protect nanoparticles, avoiding irreversible coalescence and improving their optical stability,
578
thus rendering photothermal ablation more effective. Chitosan-based coating significantly
579
changes the surface property of nanoobjects. Chitosan is an excellent agent for nanoparticle
580
biocompatibilization and stabilization, but it can also be useful as a scaffold structure for
581
anchoring the antibodies and proteins needed for selective cellular targeting.82 Thanks to the
582
presence of amino groups, chitosan can be applied to drug or DNA delivery systems, since it
583
can be ionically crosslinked to multivalent elements. Due to its properties, chitosan has
584
already been widely used for coating metal nanomaterials such as gold and silver
585
nanoparticles.83
586
Boca et al. have focused on the study of photothermal activation in chitosan-coated silver
587
nanotriangles (Chit-AgNTs) for treating human lung cancer cells (NCI-H460).82 Chit-AgNTs
588
have been prepared by triggering the reaction of silver nitrate in chitosan solution. Cells have
589
been incubated with Chit-AgNTs and irradiated by a laser at different power densities for 10
590
min at 800 nm. The Authors found that the rate of cell mortality in the presence of Chit-
591
AgNTs is higher than in the presence of thiolated poly(ethylene) glycol-capped gold
592
nanorods. Cytotoxicity tests have shown that Chit-AgNTs are efficiently uptaken by cancer
593
cells, while exhibiting good biocompatibility for healthy human embryonic cells.
594
Zhuang et al. have synthesized gold nanoparticles coated with chitosan or O-carboxymethyl
595
chitosan (CMCS), and studied them as therapeutic agents for the photothermal ablation of
596
cancer cells, such as the hepatocellular carcinoma cell line (HepG2) and human dermal
597
fibroblast cells (HDF).84 The particle functionalization procedure relies on mixing chitosan or
598
CMCS with as-synthesized gold particle suspensions. During in vitro tests, HepG2 and HDF
599
cell lines have been incubated in a medium containing CN or CMCS gold nanoparticles for
600
2 h. Cells have been exposed to 817 nm NIR laser (5 W cm-2) for 2 min. The highest 28 ACS Paragon Plus Environment
Page 28 of 84
Page 29 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
601
efficiency of the photothermal ablation has been observed for the cells treated with
602
gold/chitosan nanoparticles. Almost 100% of HepG2 and 90% of HDF cells have been killed
603
after the laser treatment.
604 605 606 607
3.3 Peptide functionalization of nanoagents
608
The diagnosis and treatment of cancer diseases at the cellular level would be greatly enhanced
609
by the ability of multifunctional nanomaterials to deliver active probes and therapeutic agents
610
into specific cells and cellular compartments. The cell nucleus is the most desired target for
611
cancer therapies. Targeted nuclear delivery of diagnostic probes and therapeutics could
612
improve disease detection and treatment. The goal of several research studies is to further
613
examine the ability of multipeptide nanoparticle conjugates to target cell nuclei. Observations
614
suggest that the most efficient nuclear targeting may result from the use of peptides attached
615
to a single vector. Combining this type of strategy with other types, such as photothermal
616
therapy, offers greater possibilities for developing effective cancer cell eradication methods.
617
Peptide layers screen the undesirable inorganic surface of nanoparticles behind an organic
618
shell, endowing the nanoagents with the needed cell-entering properties together with the
619
required optical properties.73
620
Daniels et al. have presented new approaches for the synthesis of pH Low Insertion Peptide
621
(pHLIP) and PEG-coated spherical and spiked gold nanoparticles for the enhancement of
622
radiation damage during NIR thermal therapy.85 It has been shown that pHLIP peptides are
623
excellent for targeting acidic tumors, and their use makes it possible to deliver therapeutic
624
agents specifically to the cancer cells inside tumors. The irradiation by an 805 nm laser of 29 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
625
multispiked gold nanoparticles coated with PEG and pHLIP led to a time- and concentration-
626
dependent increase in temperature. The developed pHLIP-coated nanoparticles can find
627
applications in targeted photothermal therapies of tumors.
628
The possibility of fabricating graphene oxide decorated with gold nanoparticles by using
629
thermostable antimicrobial nisin peptides as functionalizing agents has been presented by
630
Otari et al. 86 This nanocomposite has been used as an agent in photothermal therapy, showing
631
an enhanced photothermal activity compared to gold nanoparticles, thanks to the presence of
632
reduced graphene oxide (rGO). Nisin peptide has a wide range of antimicrobial activities
633
against gram-positive microorganisms. During in vitro tests, cells have been treated with the
634
synthesized nanocomposite and irradiated using an 800 nm laser (at a power density of 0.5 W
635
cm-2). The photothermal effect of nanocomposites on MCF7 breast cancer cells caused the
636
inhibition of approximately 80% of the cells after a 5 min irradiation.
637
Tan et al. have demonstrated that the lycosin-I-conjugated spherical gold nanoparticles
638
(LGNPs), lycosin-I-modified gold nanorods (LGNRs), and gold nanorods (GNRs) showed an
639
efficient cellular internalization efficiency toward cancer cells, also displaying an
640
unprecedented selectivity over noncancerous cells.87 LGNRs and LGNPs have been examined
641
as platforms for cancer photothermal therapy. In vitro tests have been carried out using HeLa
642
cells previously incubated with LGNRs or peptide-modified gold nanorods (PGNRs) and
643
near-infrared irradiation (808 nm) at a power density level of 2 W cm-2 or 5 W cm-2. In vivo
644
experiments using BALB/c nude mice with xenograft tumor were performed by intravenously
645
injecting LGNRs, PGNRs and sterile PBS three times, with a time interval of 3 days. The
646
cancer sites of each mouse have been irradiated by a 808 nm laser at power density of 3.54 W
647
cm-2 for 5 minutes at a time point of 24 and 48 hours after each injection. LGNRs exhibited
648
selective intracellular translocation toward cancer cells and an efficient photothermal effect
649
under near-infrared (808 nm) irradiation, which consequently effectively killed cancer cells in 30 ACS Paragon Plus Environment
Page 30 of 84
Page 31 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
650
vitro and in vivo. Therefore, the established LGNPs and LGNRs possessed great potential in
651
cancer-targeted delivery and photothermal therapy.
652 653
3.4 Functionalization with nucleic acids
654
The functionalization of nanoparticles with nucleic acids has many advantages, offering the
655
possibility to combine photothermal therapy with gene therapy.88 The aim of gene therapy is
656
to treat cancer by using nucleic acids to inhibit the expression of genes which drive tumor
657
progression. Small interfering RNA (siRNA) is one of the main types of nucleic acids useful
658
in gene regulation. Its function is the suppression of gene expression, which triggers the
659
degradation of RNA molecules inside the cytoplasm of cells. This way, the production of the
660
encoded disease-associated protein is stopped.89
661
Nanomaterial surface functionalization with siRNA has been used in the study presented by
662
Wang et al.
663
photothermal agent showed the ability to deliver siRNA oligos targeting the BAG3 gene,
664
which efficiently blocked the heat-shock response. Gold nanorod-siRNA agents have been
665
prepared by combining anionic-charged siRNA oligos with gold nanorods whose surface was
666
previously modified by negatively-charged poly(sodium 4-styrenesulfonate) (PSS) and
667
positively-charged
668
experiments, Cal-27 cells have been treated with functionalized gold nanorods for 24 h and
669
irradiated by a 810 nm wavelength laser at different power densities. In the case of in vivo
670
tests, mice infected with tumors have been injected with a gold nanorod-siRNA sol, and
671
irradiated by the 810 nm laser with a energy density of
672
treatment, the tumors have been extracted and analyzed (Figure 7). In vitro and in vivo studies
673
demonstrated the ability of the siRNA-functionalized nanomaterials to overcome
90
They have modified gold nanorods using siRNA and the developed
poly(diallyldimethylammonium
chloride)
31 ACS Paragon Plus Environment
(PDDAC).
In
in
vitro
600 J cm-2. 18 days after the
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
674
thermoresistance and produce the maximal tumoricidal effect with minimal damage to normal
675
tissues. The inhibition of BAG3 expression makes cancer cells more vulnerable to
676
chemotherapy or radiotherapy, thereby eliminating their therapeutic resistance and enhancing
677
therapeutic efficacy.
678 679
Figure 7. (a) Tumor growth percentage from different groups during the first 18 days after
680
Gold nanorod-siRNA treatment. (b) Photographs of tumors 18 days after G1-G5 treatments.
681
Reprinted from ref. 90. Copyright 2015, with permission from Elsevier.
682 683
Braun and co-workers have used gold nanoshells modified with siRNA for gene silencing.
684
They have used a NIR laser to control the release of siRNA from nanoshells.91 It has been
685
shown that the intense local heating by pulsed lasers causes vesicle rupture, which allows the
686
drug delivery through membranes. In order to study the system efficiency, the Authors used a
687
GFP-expressing cell line and the combination of siRNA release with nanoshell-induced
688
endosome rupture has been explored. Cells incubated with nanoshell-siRNA have been
689
irradiated by a 800 nm pulsed laser with a power density of 3.5 W cm-2. All samples showed
690
high viability, but the GFP level for laser-exposed nanoshell-siRNA showed a gene silencing
691
of about 80% . 32 ACS Paragon Plus Environment
Page 32 of 84
Page 33 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
692
The use of pulsed near-infrared laser irradiation for the release of nucleic acid from gold
693
nanoparticles has also been investigated by Takahashi et al.92 They used plasmid DNA
694
immobilized on gold nanorods modified by phosphatidylcholine (PC-GNRs). DNA-PC-GNR
695
sols under irradiation with a 1064 nm laser. The results showed that only tens of seconds of
696
NIR laser irradiation are necessary to induce DNA plasmid release. Such a rapid release is
697
very beneficial for minimizing unwanted photochemical damage.
698
Another exciting application of photothermal therapy combined with gene therapy is the
699
possibility to treat not only cancer, but even diseases caused by microbial infections. The use
700
of aptamer-functionalized gold nanoparticles (Apt@Au NPs) and gold nanorods (Apt@Au
701
NRs), for the inactivation of Methicillin-resistant Staphylococcus aureus (MRSA) by targeted
702
photothermal therapy, has been reported on by Ocsay et al.93 The DNA aptamer was
703
specifically selected to MRSA cells and used for targeted therapy. Au NRs and Au NPs acted
704
as convertors for transducing NIR radiation to heat for the photothermal inactivation of cells.
705
Mixtures of aptamer-functionalized nanoparticle or nanorod sols and the cell solution were
706
incubated for 1 h, and then illuminated using a single-mode NIR laser with 808 nm
707
wavelength at 1.1 W cm-2 power density. Results showed that both Apt@Au NPs and
708
Apt@Au NRs specifically bound to MRSA cells, but only Apt@Au NRs effectively
709
inactivated MRSA cells through hyperthermia due to their longitudinal absorption of NIR
710
radiation and photothermal conversion. The Authors also observed that the MRSA Apt@Au
711
NRs are very effective and promising nanosystems for specific cell recognition and in vitro
712
PTT. Au NRs have multiple functions, serving as a nanoplatform for aptamer immobilization
713
and also providing multivalent effects for increasing the binding strength of aptamer for the
714
targeting of MRSA cells; moreover, they act as photothermal agents to inactivate cells with
715
the heat generated under NIR irradiation.
33 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
716
The functionalization of nanoparticles used in photothermal therapy offers the possibility to
717
combine this therapy with other innovative therapeutic methods, such as chemotherapy,
718
immunotherapy, and gene therapy. The use of appropriate modifications not only increases
719
the therapeutic effectiveness, but also prevents damage to healthy cells and tissues. The
720
studies described clearly demonstrate the great efficiency of polymer-functionalized
721
nanoparticles in killing cancer cells. Some recent studies, however, have shown that
722
photothermal therapy is always accompanied by a heat shock response. This effect shields
723
cancer cells from hyperthermia damage, consequently decreasing its therapeutic efficacy.
724
Usually, to increase therapeutic efficacy, a larger administration dose or higher irradiation
725
power is necessary, causing severe side effects due to the overheating of neighboring normal
726
tissues. The use of gene therapy combined with phototherapy is a fascinating candidate for
727
solving this problem. Killing cancer cells without inducing cellular thermoresistance by
728
photothermal therapy is one of the biggest challenges in this field.
729 730
34 ACS Paragon Plus Environment
Page 34 of 84
Page 35 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
731
4. Polymer Matrix Composites
732
Polymer matrix composites are materials formed from at least two different types of
733
components, e.g. polymer matrix and ceramic/metal nanofillers.94 In these materials, the
734
polymer matrix is a hosting network, while nanofillers are the photothermal agents. The
735
objective of composite development is to create new materials which will combine the
736
advantages of both components at the same time. Today, the latest trend of cancer treatment is
737
the development of techniques based on a combination of various treatment methods (e.g.
738
combined chemo- and photothermal therapy) which operate locally. Local treatments make it
739
possible to maximize the bioactive molecule concentration in the cancer area, while
740
minimizing the drug toxicity for other tissues. This type of treatment has been considered
741
much more effective than conventional chemotherapy.95
742
Various delivery systems such as drug-loaded fibers or hydrogel have been studied for local
743
therapy in recent years.96,97 As we have already pointed out, however, a single cancer
744
treatment method is not sufficient, because of both the inefficient drug permeability in the
745
cancer site, and the drug resistance of tumor cells.98 Therefore, thermally responsive polymer-
746
matrix composites are a promising solution because they can easily host drugs and
747
photothermally-active particles. Moreover, because of the polymer responsivity, the final
748
material can be used as an on-off switch for drug release under NIR irradiation.99 In the case
749
of drug release, the hyperthermia effect of photothermal therapy not only kills cancer cells,
750
but also increases drug penetration into the tumor site.100,
751
caused by the non-target accumulation occur, together with an unsatisfactory anticancer
752
efficiency.100 However, drug release is still a not entirely controllable process. Furthermore,
753
additional advantages of the use of polymers in biomedical applications stem from the
754
biodegradable nature of these materials.102
101
35 ACS Paragon Plus Environment
In this case, the side effects
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
755
The properties of polymer composites (e.g. mechanical properties, degradation rate, and drug
756
release) are highly dependent on structural morphology at the molecular level, such as
757
crystallinity fraction or crystallite size, and polymer chain structure. It is worth remembering
758
that polymer structure is characterized by temperature-dependent features. When surface
759
plasmon resonance (SPR) is induced in the polymer composite, due to the inclusion of
760
inorganic nanofillers, energy is transferred from the particle to the surrounding polymer
761
matrix, causing local polymer melting and, consequently, leading to changes in its structure.94,
762
103-105
763
described by Viswanath et al. 103 They demonstrated that a low annealing temperature reached
764
during the photothermal process led to the maximum polymer crystallinity in a shorter
765
annealing time than with conventional annealing procedures (Figure 8). Gold nanoparticles
766
embedded into polymer matrixes are localized heat sources which make it possible to
767
manipulate crystallinity, crosslinking, or chemical reactions. Using this nature-inspired
768
cascade approach, the internal structure of polymer matrix composites can be altered at any
769
point in their life cycle by photothermal treatments. Indeed, light responses can act as
770
mediator signals which generate material structural transformations and, in turn, offer
771
additional functionalities (e.g. on-off release of bioactive molecules) to the material.99, 102, 106
The effect of photothermal heating on the polymeric structure has been extensively
772
36 ACS Paragon Plus Environment
Page 36 of 84
Page 37 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
773 774
Figure 8. Images of poly(ethylene oxide) (PEO) spherulites observed through a cross-
775
polarized optical microscope of: (a) melt-crystallized gold nanoparticle:PEO film; (b, c, d)
776
after conventional annealing at 60 °C and after photothermal process; (e, f, g) at different time
777
points; (h) crystallinity; (i) spherulite density as a function of annealing time for gold
778
nanoparticle:PEO melt-crystallized films. The annealing was conducted by conventional and
779
photothermal heating. Reprinted with permission from ref. 103. Copyright 2013 American
780
Chemical Society.
781 782
Today there are only a few scientific papers devoted to polymer matrix composites. They are
783
briefly described in the following paragraphs.
784
37 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 38 of 84
785
4.1 Hydrogels
786
Hydrogels are three-dimensional constructs formed by hydrophilic polymers, in which the
787
polymer chains are crosslinked physically or chemically to form a network. The capability to
788
accumulate large amounts of water is a characteristic feature for hydrogels, which may reach
789
up to 90% of their bulk mass. High water content, softness, flexibility, and biocompatibility
790
make hydrogels very similar to natural tissues. At the same time, this type of material has
791
certain limitations, e.g. toxic crosslinking agents which may remain in the bulk,
792
inhomogeneity of nanoparticle dispersion, or poor mechanical properties.107 Moreover, a few
793
hydrogels are responsive to different kinds of environmental condition changes, like pH or
794
temperature. Thermosensitive hydrogels are characterized by lower and upper critical solution
795
temperatures (LCST, UCST), which indicate the temperature points of hydrogel state
796
transitions. Some hydrogels formed for controlled drug release show a LCST slightly above
797
body temperature. When the temperature of the copolymer exceeds the LCST, the hydrogel
798
collapses and releases any components suspended in the hydrogel matrix.107-109 Hydrogels are
799
currently used for manufacturing various medical devices, such as contact lenses, tissue
800
engineering scaffolds, drug delivery systems, and wound dressings.107 The application of this
801
type of material for photothermal cancer therapy was recently explored by several scientists.99,
802
110-112
803
Hydrogel materials for cancer therapy have been developed and studied by Conde et al.
804
These Authors proposed the use of one single hydrogel patch capable of combining gene,
805
drug, and photothermal therapies all at the same time. This hydrogel contains nanofillers such
806
as gold nanorods and gold nanospheres. Nanorods have been functionalized with appropriate
807
biomarkers, in order to enhance their uptake by cancer cells. These biomarkers made it
808
possible to eliminate the problem connected with the absorption by healthy cells and the
809
strong hypothermia impact on them. Using confocal microscopy and proper labeling 38 ACS Paragon Plus Environment
99
Page 39 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
810
techniques, the Authors proved that nanoparticles are more effectively absorbed by CRC cells
811
(cancer cells) than 3T3 fibroblasts. MTT assay also confirmed the decreased tumorgenicity of
812
CRC cells after laser irradiation. The hydrogel formed using poly(amidoamine) and the
813
abovementioned particles was analyzed in in vivo tests using mouse models (Figure 9). It has
814
been shown that the designed hydrogel nanocomposite is a good material for nanoparticle and
815
drug delivery into cancers, as well as an effective carrier for the quick diffusion of therapeutic
816
agents into cancer cells. However, some questions concerning implantation procedure (e.g.
817
depth, insertion, and fixation) still remain under debate. Moreover, the rapid changes in the
818
material structure and its consequent possible damage after reaching the LCST point, which
819
facilitates the quick release of drugs, remains an unsolved problem.
820 821
Figure 9. (a) Schematic of the smart hydrogel–nanoparticle patch with drug–siRNA
822
nanoparticle conjugates (drug–gold nanorods and siRNA–gold nanospheres) for local gene
823
and drug release designed by Conde et al.99. (b) Fluorescence and thermographic images of
824
hydrogel patches. (c) Thermographic surveillance of the photothermal heating of hydrogel
39 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 40 of 84
825
patches in mice (nD5) 72 h after the first NIR treatment. Reprinted by permission from
826
Springer
827
http://www.nature.com/nmat/
Nature,
ref.
99.
Copyright
2016,
Macmillan
Publishers
Limited.
828 829
In order to improve the implantation of hydrogel composites, injectable materials should be
830
taken into consideration. Xing et al. have described an injectable, self-healing hydrogel with
831
self-assembly biomacromolecules.110 In this study, natural polymers such as collagen, elastin,
832
or silk fibroin have been used. The interactions between polymer gelators and nanoparticles
833
have been used to assemble tunable and self-healing polymeric materials. Additionally,
834
nanoparticles play a key role in tuning mechanical properties, while the polymeric matrix
835
remains a good environment for water-soluble drug diffusion. The Authors extensively
836
described the healing mechanism of this hydrogel, focusing on the gold nanoparticle-collagen
837
interaction. It has been proven that intratumoral-injected hydrogel sustained in situ and that
838
the drugs were successfully released.
839
A few studies reported that the process of selective treatment of cancer cells by hyperthermia
840
needs to be improved.111 The hydrogel composite formed with spinach extract, reduced
841
graphene oxide, gold nanocages (AuNCs), and fluorouracil (5-FU) effectively increased the
842
temperature and released the drug, leading to a successful reduction in HeLa cell proliferation.
843
But as it was shown, CHO cells, used as a control, also exhibited a reduced proliferation. This
844
effect is due to the rGO and/or AuNC release, which additionally affects CHO cell
845
proliferation. The Authors, however, only presented profiles for 5-FU, without showing data
846
on rGO or AuNCs release.111 In this case, the effectiveness and targeting ability of the active
847
compound could be modified by appropriate biomarkers as proposed by Conde et al. 99
40 ACS Paragon Plus Environment
Page 41 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
848
For hydrogels, as well as for any other type of 3D materials, implantation remains a crucial
849
problem. A good idea for the proper placement of hydrogel composite components is the in
850
situ crosslinking. Zhang et al. have explored this type of strategy with a view to synthesize a
851
novel hydrogel for synergistic cancer therapy.113 The hydrogel has been formed by
852
poly(ethylene glycol) double acrylates (PEGDA), a titanium dioxide/multiwall carbon
853
nanotube (TiO2@MWCNT) nanocomposite, which acts as a photoinitiator and photothermal
854
agent for cancer multi mechanism therapy, while DOX has been applied as a model drug.115
855
The proposed material showed a certain recyclability, which may be important for successful
856
treatments, and a long-lasting drug release. However, the reported data also showed that
857
limited UV or NIR light penetration may be an additional problem. Because of these reasons,
858
this material may not be suitable for deeply located tissue treatments.114, 115
859
An injectable drug delivery system which provides photothermal transduction activity was
860
proposed by Zhou et al.
861
(SWNTs), PNIPAM hydrogel, and doxorubicin. The combination of hyperthermia therapy
862
and controlled DOX release gave a higher cancer suppression rate on gastric cancer xenograft
863
mouse models than did free DOX, and without detectable organ toxicity. However, the
864
application of rGO or carbon nanotubes in medical treatments seems to be yet uncertain. The
865
release profiles, accumulation in tissues, and cytotoxicity are constantly studied, and at the
866
same is still unclear.117, 118
867
Polymer matrix composites in the form of hydrogels may be successfully used as medical
868
materials for combined cancer therapy (chemo- and photothermal therapy). Some problems
869
concerning the nature of these materials should also be solved. Furthermore, the accurate
870
control of the heating under NIR irradiation to avoid damaging the hydrogel structure and the
871
fast drug release will be the biggest challenge in this field.
116
The developed system contains single-wall carbon nanotubes
41 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
872
4.2 Fibrous membranes
873
Polymer membranes already play a significant role in fields such as water purification, fuel
874
cells, cell scaffolds, and wound dressings. Membranes may be formed by various techniques
875
e.g. melt electrospinning 119 or electrospinning,120-124 solvent casting and particles leaching 125
876
or freeze-drying.126 In the case of polymer matrix composites for photothermal cancer therapy,
877
however, just a few of these have been investigated. Nonwovens can be fabricated by various
878
techniques, thus making it possible to obtain fibers with diameters ranging from dozens of
879
nanometers to a few micrometers.120-124 Fibrous nanomaterials are promising materials for
880
medical applications such as photothermal cancer therapy, due to their highly active surface
881
area, to which drugs or particles can be attached.
882
The concept of using fibers for photothermal therapy is quite new, and only a few papers have
883
been published recently on this subject. Zhang et al. proposed poly-L-lactic acid (PLLA)
884
composite nanofibers loaded with multiwalled carbon nanotubes (MWCTs) and
885
doxorubicin.102 Moreover, in this scientific work, polytherapy (chemo- and photothermal
886
therapy) has been suggested for cancer treatment and the on-off switching mechanism of drug
887
release was described. PLLA has a low glass-transition temperature (Tg) of around 60°C.
888
Since the human body temperature is under the Tg of PLLA, the as-prepared material is in its
889
glassy state and the polymer chain mobility is limited. Therefore, the release of DOX from the
890
fibers is rather slow (5-8% per 12 h, depending on pH). After irradiation, however, a great
891
deal of heat is efficiently generated by MWCNTs. When the temperature exceeded the PLLA
892
Tg, the mobility of the polymer chain increased, enhancing the drug release (20% per 2 h after
893
30 minutes of irradiation). This work proved that nanofibers can effectively combine two
894
cancer treatment methods, while delivering drugs effectively and locally. This material can be
895
easily used for the treatment of skin cancer, but its implantability remains uncertain.
42 ACS Paragon Plus Environment
Page 42 of 84
Page 43 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
896
Furthermore, as was described in the previous paragraph, the problem connected with the
897
MWCNT cytotoxicity is still unsolved.
898
Nonwovens for photothermal therapy were also analyzed by Shao et al.127 Locally produced
899
energy was an effect of the bismuth selenide (Bi2Se3) irradiation. In this work, Bi2Se3
900
nanoplates were incorporated into the poly(lactide) fiber matrix and tested as photothermal
901
agents (Figure 10).
902 903
Figure 10. (a) Scanning electron microscope (SEM) and transmission electron microscopy
904
(TEM) images of PLLA and Bi2Se3/PLLA membranes. (b) Infrared thermographic maps and
905
time-dependent temperature increase in the tumor-bearing nude mice irradiated by an 808 nm
906
laser. (c) Temperature growth during the irradiation of PLLA and Bi2Se3/PLLA membranes.
43 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
907
(d) Corresponding growth curves of tumors in different groups of mice after NIR laser
908
irradiation. Reprinted with permission from ref. 127. Copyright 2017 American Chemical
909
Society.
910 911 912
The Authors showed that composite fibers effectively increase the temperature of the
913
medium, while killing HeLa cancer cells. In this case, however, one important issue should be
914
further taken into consideration: the Authors did not present data concerning bismuth selenide
915
release from fibers before and after irradiation, nor on its cytotoxicity for non-cancer cells. If
916
such a significant temperature increase appears during irradiation, PLLA molecule mobility is
917
increased and nanoplates may be easily released to the environment.128 This problem is not
918
visible when analyzing monocultures of HeLa cells, because target cells are logically
919
successfully killed. However, in co-culture, such as in in vivo conditions, both healthy and
920
cancer cells coexist. In this case, it is hard to say if either only local hypothermia is
921
responsible for cell death or if the cytotoxicity of bismuth selenide is also a cause of it.127 The
922
concept of selectively attaching biomarkers could be used in this case, as was done by Conde
923
et al. 99 to limit bismuth selenide effects on healthy cells.
924
Additionally, nanofibers may be surface-functionalized in order to introduce bioactive
925
components for treatment purposes.129 However, the attached substances should be carefully
926
selected in order to avoid inducing cytotoxic effects.118 Moreover, it should be remembered
927
that surface-functionalized fibers show very different particle/drug release kinetics and
928
photothermal effect efficiency than do bulk loaded fibers.
929
Various technologies are available today to fabricate fibers on a mass scale. Most of them use
930
polymer solutions or melted polymers. This may be problematic if biologically active 44 ACS Paragon Plus Environment
Page 44 of 84
Page 45 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
931
molecules for combined therapy are to be added to fibers. Aggressive solvents or high
932
temperature could cause the degradation of these components. To summarize, the
933
implantation procedure of nonwovens, their degradation in in vivo conditions, and the
934
influence of solvent traces should be further investigated.
935
Only a few scientific articles on polymer nanocomposite membranes with different
936
morphologies for photothermal therapy have been published recently.130-132 Li et al. fabricated
937
polydimethysiloxane (PDMS) membranes containing gold nanoparticles which have been
938
synthesized by the in situ method.130 In the case of composite membranes with nanoparticles,
939
the potential segregation of components may occur, thus leading to a higher intensity of the
940
NIR radiation needed for photothermal effects. To avoid this problem, Li et al. decided to use
941
in situ polymerization. Wet and dry samples of the proposed composites have been studied. It
942
has been demonstrated that the amount of remaining solvent traces affects the intensity of the
943
photothermal effects. Upon laser irradiation, wet membranes reached lower temperatures than
944
dry membranes. The Authors explained that, most likely, the heat generated by gold
945
nanoparticles is dissipated throughout the entire system (both the remaining solvent and the
946
polymer matrix) in wet membranes, resulting in a lower temperature of these materials
947
compared to dry membranes.131, 132 The data presented may help to understand the differences
948
in the efficiency of photothermal effects in the previously mentioned hydrogels or nonwoven
949
materials.
950
Up to this time, various forms of polymer matrix composites for photothermal cancer
951
treatment have been investigated. As can be seen, all of them show significant advantages, i.e.
952
are good vehicles for drugs and nanoparticles, and induce localized hyperthermia, but they
953
also have a few disadvantages: a not fully controlled drug release and a limited implantation
954
localization and depth. In spite of this, polymer matrix composites are the most promising
45 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
955
materials for the development of functional materials for the combined chemo- and
956
photothermal therapy of cancer.
957 958 959
5. New Trends
960
In the fight against cancer, compared with traditional therapeutic approaches (e.g.
961
chemotherapy, radiotherapy and immunotherapy), PTT exhibits very unique advantages such
962
as selective targeting and minimal invasiveness. PTT has the capability to destroy cancer cells
963
in the primary tumor (mainly if localized in superficial tissues) and to combat the initial stage
964
of cancer metastasis. Moreover, PTT can effectively destroy cancer stem cells and tumor
965
initiating cells avoiding metastatic propagation to distant organs. It is important to say that
966
PTT can kill cancer cells via cellular necrosis, enabling an inflammatory responses that can
967
affect the efficacy of the same treatment. To overcome this issue, it has been demonstrated the
968
possibility to trigger the PTT heat in such a way to induce apoptosis (does not generate an
969
inflammatory process) rather than necrosis.10 On the other hand, at the current stage PTT fails
970
if applied after the metastatic process has taken place, such as in presence of aggressive
971
tumors involving important organs or tissues which are already far away from the primary
972
tumor. To overcome this issue, novel nanoplatforms able to efficiently combine PTT with
973
other therapies to exert a stronger impact in presence of metastasis has been combines. In this
974
framework, photoresponsive nanoagents have been exploited for combined PTT-
975
immunotherapy.133 Results have shown that nanotubes-based PTT could modulate the
976
adaptive immune responses for the treatment of metastatic cancers. PTT agents could not only
977
be used for photothermal tumor destruction but also act as immunological adjuvants
978
promoting the maturation of dendritic cells and production of anti-tumor cytokines. These
46 ACS Paragon Plus Environment
Page 46 of 84
Page 47 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
979
data prove that after photothermal ablation of the primary tumor, the induced immunological
980
responses are able to inhibit a secondary tumors as well as lung metastasis. Another approach
981
is based on the fact that, cellular dysfunctions due to radiation are mediated by the
982
microvascular endothelial damage and that therefore chemical vascular disrupting agents
983
could be effective in combination with radiation therapy for the tumor vascular disruption.
984
The use of these agents are promising materials to improve therapeutic outcome and
985
extending their applicability to tumors that barely respond to standard therapies. Kunjachan et
986
al. proposed a dual-targeting strategy to improve tumor blood vasculature radiation outcome
987
by inducing vascular damage.134 This study, experimentally validated the hypothesis that
988
tumor-specific vascular disruption could be mediated by the administration of targeted
989
nanoparticle followed irradiation which could be easily extended to PTT nanoplatforms.
990
The development of novel nanomaterials for photothermal therapies is evolving rapidly.
991
Current strategies concern not only monotherapies such as the NIR-controlled heating of
992
cancerous cells with, for example, gold nanorod nanoparticles. The novel advanced medical
993
treatments require the development of combined therapies using NIR illumination for
994
inducing chemotherapeutic drug release or photodynamic therapy. One of the emerging
995
techniques used for the formation of multifunctional biomaterials is electrospinning. In this
996
method, a polymer solution is stretched by electrostatic forces which develop between a
997
needle, connected to a high voltage supply, and a grounded collector in order to form ultra-
998
thin fibers. Moreover, due to the nanosized fiber diameter, the developed materials show
999
structural similarity to the naturally produced proteins and polysaccharide complexes present
1000
in the extracellular matrix (ECM), positively influencing cellular proliferation and activity.121
1001
As we will show in this paragraph, electrospinning can combine all the properties of the
1002
abovementioned materials, conjugated polymers can be electrospun, while the electrospun
1003
nanomaterial surface can be easily functionalized, and it is even possible to fabricate 47 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1004
composite nanomaterials to develop novel heat generating devices. One of the best examples
1005
of this is the development of CaTiO3:Yb,Er nanofibers co-conjugated with Rose Bengal (RB)
1006
and gold nanorods.135 The incorporated RB served as the photodynamic therapy agent, while
1007
the gold nanorods attached to the nanofiber surface were responsible for the hyperthermia
1008
effect. Additionally, upconversion photoluminescence (UCPL) of CaTiO3:Yb,Er nanofibers
1009
was used to excite RB and gold nanorods, so photodynamic and photothermal therapy could
1010
occur simultaneously, during the irradiation with a laser source (λ=980 nm) with a deeper
1011
tissue penetration. The combination of several treatments is particularly significant due to the
1012
resulting synergistic effect that shrinks the tumors and stops the metastatic spread of cancer,
1013
in which cells escape from the primary tumor and form new ones. Moreover, every
1014
monotherapy has its own side effects; therefore, combining different therapeutic approaches
1015
can help eliminate heterogeneous cancer cells that cannot be treated with single
1016
chemotherapeutic agents due to drug resistance.
1017
The rise of new strategies in the treatment of tumors is essential for the complete eradication
1018
of cancer cells from the body. PTT is a promising method for increasing temperature to a
1019
level that induces the apoptosis of cancer cells via localized hyperthermia; on the other hand,
1020
this method also makes it possible to trigger other processes that could also help eliminate
1021
cancer cells. One of these methods is the on-off release of drugs. For instance, Park et al. have
1022
described an experiment in which fibrous polycaprolactone (PCL) materials containing phase-
1023
changeable fatty acids and indocyanine green were illuminated with NIR light.136 Upon NIR
1024
illumination, the temperature of the material increased above the melting point of the fatty
1025
acids, thus causing a greater drug release from the material. This strategy could limit the
1026
harmful impact of chemotherapeutic drugs, being more beneficial for patients’ health.
1027
Another interesting functionality that could help clinicians in assessing the response to
1028
treatment is the monitoring of the drug release from the implanted biomaterial itself.137 The 48 ACS Paragon Plus Environment
Page 48 of 84
Page 49 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1029
incorporation of Yb3+/Er3+ into CaTiO3 nanofibers helped control the release of doxorubicin;
1030
additionally, the amount of this kind of bioactive molecule could be easily assessed optically
1031
by evaluating the intensity ratio of green to red emissions of UCPL nanofibers under
1032
irradiation with a 980 nm laser. Another innovative strategy is the simultaneous capture and
1033
killing of cancer cells described by Mauro et al.138 They have functionalized the surface of
1034
PCL fibers with graphene oxide. This allowed the selective capture of circulating tumor cells
1035
and upon irradiation of NIR light, the temperature increase eradicated them photothermally.
1036
This is a potential tool not only for eliminating metastatic cells, but also for the early detection
1037
of cancer cells thanks to the fluorescence properties of this material. In any case, while the
1038
idea of capturing and eradicating circulating tumor cells with NIR light is promising, the long-
1039
term consequences of using graphene oxide in implantable nanomaterials are not well known.
1040
The abovementioned hybrid scaffold could be implanted under the patient’s skin, thus making
1041
it more accessible for NIR light illumination. However, new PTT agents are necessary for
1042
treating inaccessible tumors with light in the NIR I region.
1043
As mentioned in the previous paragraph, numerous photothermal agents, especially
1044
nanomaterials with strong absorbance in the tissue-transparent NIR I (650-950 nm), region
1045
have been studied extensively for use in PTT. The low penetration depth of laser light is one
1046
of the limiting factors which permit treating superficial cancers only, such as skin and breast
1047
tumors. The illumination of tissues by lasers operating in the NIR II window (950-1700 nm)
1048
offers a higher light penetration depth. One of the most fascinating examples of this is the use
1049
of CaTiO3:Yb,Er nanofibers which, thanks to the UCPL, emit light-activating photosensitizers
1050
and heat-generating agents, leading to a synergistic effect of photodynamic and photothermal
1051
therapies, while irradiated with a 980 nm laser.135, 137 Nanomaterials filled with nanoagents
1052
which absorb light in the NIR II region, have a high aspect ratio, and present upconversion
49 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 50 of 84
1053
photoluminescence could potentially be used for the formation of theranostic medical devices,
1054
opening new possibilities for the simultaneous diagnosis and efficient treatment of tumors.
1055
6. Conclusions and Future Perspectives
1056
From the very beginning, inspiration from nature has proved to be very helpful for the
1057
development of advanced materials with novel and unpredictable properties, looking at the
1058
mechanisms involved in life processes as they occur in nature, but with an in-depth
1059
knowledge of material sciences. Proper design and application of theranostic biomaterials
1060
based on polymer PTT agents require a deep understanding of the features that distinguish
1061
them from other smart materials. Polymer-based PTT nanoplatforms offer many advantages,
1062
such as ease of fabrication, biocompatibility and especially the possibility to tune their
1063
cascaded responsive properties in order to trigger other local cancer treatments such as
1064
chemo-, gene-, or photodynamic therapy.
1065
This review provides an overview on the progress made towards the production of different
1066
types of polymer-based multifunctional nanomaterials for PTT. Three main material classes
1067
have
1068
nanoparticles and polymer matrix composites. All of them are composed by different types of
1069
polymers and the processes used in responding to light stimulation in a controlled cascade-
1070
like fashion are based on different mechanisms, therefore every described class presents
1071
specific advantages and shortcomings.
1072
In summary, conjugated polymers are highly versatile materials which can be synthesized
1073
through well-established reactions (e.g. click chemistry, copolymerization, ester-coupling and
1074
polycondensation). Moreover, they can be functionalized after polymerization in order to
1075
introduce novel functional groups. This makes these polymers ideal building blocks for
1076
materials with enhanced light-to-heat conversion (e.g. improving the light-harvesting ability
been
identified:
conjugated
polymer
nanomaterials,
50 ACS Paragon Plus Environment
polymer-functionalized
Page 51 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1077
in the second NIR window). Additionally, conjugated polymers such as polythiophene
1078
derivatives can be easily modified in order to increase their water solubility. Conjugated
1079
polymers can be modified to finally form copolymers with D-A properties and they can be
1080
combined with wide-absorption organic dyes to further increase their photothermal
1081
conversion efficiency. Conjugated polymers in the form of nanoparticles can help control the
1082
drug release process; they can also improve the treatment efficacy and, at the same time,
1083
could minimize the side effects of, for example, chemotherapies. Moreover, nanoparticles, in
1084
addition to transporting a drug cargo, can also be bound to target cells and generate signals for
1085
imaging purposes as well as generating ROS to perform PTT and PDT simultaneously.
1086
However, several challenges need to be addressed before these nanosystems can find a
1087
clinical application: low photothermal conversion efficiency and low photostability in vivo,
1088
for instance. Another issue is the fast renal clearance of nanomaterials and their over-
1089
accumulation in non-targeted tissues, which may trigger additional serious side effects.
1090
Therefore, we can conclude that platforms based on conjugated polymers have shown
1091
outstanding light-material interaction properties as well as exceptional multifunctional
1092
performance, but the interaction of these materials with body tissues should be the subject of
1093
further investigations before they can be considered as biomaterials suitable for potential
1094
applicability to clinical oncology.
1095
The surface functionalization of PTT nanoparticles by polymers was initially explored in
1096
order to stabilize nanomaterials and to increase their biocompatibility prolonging their
1097
circulation within the body and avoiding the direct contact of body tissues with the metal or
1098
inorganic surface of the particles. Recently, nanoparticle capping by biomolecules such as
1099
peptides or nucleic acids was used to create multifunctional platforms able to combine PTT
1100
with other advanced cancer therapies (e.g. gene therapy) activated by the interaction of the
1101
nanomaterials with the NIR radiation. This intriguing methodology created new opportunities 51 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1102
in the biomedical field. However, it is worth pointing out that the presence of functional
1103
polymer layers around PTT nanoagents does not allow a significant improvement of the
1104
efficiency of the photothermal effect, since it is mainly driven by the core features.
1105
Polymer matrix composites are the last class of nanomaterials that has been explored in this
1106
well-detailed overview. These materials have been investigated in depth only in the last
1107
decade, but their scientific interest is growing quickly, because of the possibility to combine
1108
the advantages of both aforementioned material classes. Therefore, in this case it is possible to
1109
improve both light-material interaction and biocompatibility as well as obtaining several new
1110
functionalities driven by the polymer matrix modification (e.g. crystallinity) which occurred
1111
during heat generation. Noteworthy, PTT hydrogel-based nanoplatforms, because of their
1112
intrinsic high water content, softness and flexibility (very similar to natural tissues), have
1113
great potential for the development of fully biocompatible multistimuli-responsive and
1114
versatile theranostic nanomedicine platforms. Most importantly, polymer composites can be
1115
easily processed by electrospinning in order to form nanofibrous platforms, paving the way to
1116
the development of tissue-engineered scaffolds and localized smart drug delivery systems,
1117
which is the most valuable perspective in the field. Today the incorporation of drugs with
1118
photothermally active agents in the polymer nanofiber matrix leads to distinct and synergistic
1119
chemo-photothermal therapies while, at the same time, providing an ideal environment for
1120
tissue regeneration after cancer removal. The use of nanocomposites for this purpose is still in
1121
the initial stages, therefore accurate pharmacokinetics and pharmacodynamics data are still
1122
unavailable. Consequently, systematic and rigorous in vivo evaluations, especially regarding
1123
the degradability, toxicity and long term side effects of nanomaterials, are needed; but they
1124
have not yet been thoroughly performed because of the complexity of the processes of
1125
nanomaterial biodistribution and elimination from the body.
1126 52 ACS Paragon Plus Environment
Page 52 of 84
Page 53 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1127
Author Information
1128
Corresponding Author
1129
*E-mail:
[email protected] 1130
Author Contributions
1131
The manuscript was written through contributions of all authors. All authors have given their
1132
approval to the final version of the manuscript.
1133
Notes
1134
The authors declare no competing financial interest.
1135
Acknowledgment
1136
This
1137
2015/19/D/ST8/03196.
work
was supported
by the National Science Centre (NCN) grant no.
1138 1139
Abbreviations
1140
WHO, World Health Organization; PTT, photothermal therapy; NIT, near-infrared; CPs,
1141
conjugated polymers; PA, photoacustic; PDT, photodynamic therapy; ROS, reactive oxygen
1142
species; ED, electron-rich; EA, electron-deficient; NPs, nanoparticles; PT, photothermal
1143
photodynamic; PANI, Polyaniline; EB, emeraldine base; ES, emeraldine salt; PNIPAM ,
1144
poly(N-isopropylacrylamide); dPG, dendritic polyglycerol; DBSA, n-dodecylbenzenesulfonic
1145
acid; DPPC, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine; FA, folic acid; DOX, doxorubicin;
1146
PPy, polypyrrole; Astx, astaxanthin; BSA, bovine serum albumin; DSPE-PEG2000, 1,2-
1147
distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]; PTh,
1148
polythiophene; ICG, indocyanine green; OSCC, oral squamous cell carcinoma; CPNs,
1149
conjugated polymer nanoparticles; DPPV, poly{2,2′-[(2,5-bis(2-hexyldecyl)-3,6-dioxo-
1150
2,3,5,6-tetrahydropyrrolo[3,4-c]pyrrole-1,4-diyl)-dithiophene]-5,5′-diyl-alt-vinylene};
1151
PtTFPP, Pt(II)-meso-tetra(pentafluorophenyl)porphyrine; PFO, poly(9,9-di-n-octylfluorenyl53 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1152
2,7-diyl);
PFBT,
poly[(9,9-dioctylflurenyl-2,7-diyl)-co-(1,4-benzo-{2,1’,3}-thiadiazole)];
1153
TPP, tetraphenylporphyrin; Cy, Cyanine; CyP, poly(heptamethine) Cy-containing derivative;
1154
BTPBF, bis(5-oxothieno[3,2-b]pyrrole-6-ylidene)benzodifurandione; BT, 3,3’-didodecyl-
1155
2,2’-bithiophene; cRGD, cyclic arginine-glycine-aspartic acid; PFVBT, poly[9,9-bis(2-(2-(2-
1156
methoxyethoxy)
1157
PIDTTTQ,
1158
2,7-diyl)-alt-co-4,9-bis(thiophen-2-yl)-6,7-bis(4-(hexyloxy)phenyl)-thiadiazolo-quinoxaline];
1159
TBDOPV, thiophene-fused benzodifurandione-based oligo(p-phenylenevinylene); DT, 2,2′-
1160
bithiophene;
1161
fluoren-2-yl)thiophene; BBT, benzo-bis-thiadiazole; BIBDF, bis(2-oxoindolin-3-ylidene)-
1162
benzodifuran-dione; BT, bithiophene; BDT-IID, 4,8-bis[5-(2-ethylhexyl)thiophen-2-yl]-2,6-
1163
bis(trimethylstannyl)benzo[1,2-b:4,5-b′]dithiophene-6,6′-dibromo-N,N′-(2-
1164
ethylhexyl)isoindigo; PSMA, poly(styrene-co-maleic anhydride); Pdots, polymer dots; DPP-
1165
DT, diketopyrrolopyrrole-dithiophene; PF-DBT5, poly[(9,9-dioctylflurenyl-2,7-diyl)-co-(4,7-
1166
di-2-thienyl-2,1,3-benzothiadiazole)]; PBdots, polymeric blend dots; PFDHTBT, (poly[(9,9-
1167
dioctylflurenyl-2,7-diyl)-co-(4,7-bis(4-hexylthiophen-2-yl) -2,1,3-benzothiadiazole)]; PEG,
1168
poly(ethylene glycol); Chit-AgNTs, chitosan-coated silver nanotriangles; CMCS, O-
1169
carboxymethyl chitosan; HepG2, hepatocellular carcinoma cell line; pHLIP, pH Low
1170
Insertion Peptide; rGO, reduced graphene oxide; LGNPs, lycosin-I-conjugated spherical gold
1171
nanoparticles; LGNRs lycosin-I-modified gold nanorods; GNRs, gold nanorods; PGNRs,
1172
peptide-modified gold nanorods; siRNA, small interfering RNA; PSS, poly(sodium 4-
1173
styrenesulfonate); PDDAC, poly(diallyldimethylammonium chloride); PC-GNRs, gold
1174
nanorods modified by phosphatidylcholine; Apt@Au NPs, aptamer functionalized gold
1175
nanoparticles; Apt@Au NRs, aptamer functionalized gold nanorods; PEO, poly(ethylene
1176
oxide); LCST, lower critical solution temperature; UCST, upper critical solution temperature;
ethoxy)-ethyl)
fluorenyldivinylene]-alt-4,7-(2,1,3-benzothiadiazole);
poly[(4,4,9,9-tetrakis(4-(octyloxy)phenyl)-4,9-dihydros-indacenol-dithiophene-
BBT-2FT,
benzo[1,2-c;4,5-c’]bis[1,2,5]thiadiazole-4,7-bis(9,9-dioctyl-9H-
54 ACS Paragon Plus Environment
Page 54 of 84
Page 55 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1177
AuNCs, gold nanocages; 5-FU, fluorouracil; PEGDA, poly(ethylene glycol) double acrylates;
1178
TiO2@MWCNT, titanium dioxide/multiwall carbon nanotube; SWNTs, single wall carbon
1179
nanotubes; PLLA, poly-L-lactic acid; MWCTs, multiwalled carbon nanotubes; Tg, glass
1180
transition temperature; Bi2Se3, bismuth selenide; SEM, scanning electron microscope; TEM,
1181
transmission electron microscopy; PDMS, polydimethysiloxane; ECM, extracellular matrix;
1182
RB, Rose Bengal; UCPL, upconversion photoluminescence; PCL, polycaprolactone.
1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197
55 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 56 of 84
1198 1199 1200
References
1201
1.
724.
1202 1203
2.
3.
4.
Glazer, E.S.; Curley, S.A. The Ongoing History of Thermal Therapy for Cancer. Surg. Oncol. Clin. N. Am. 2011, 20, 229-235.
1208 1209
Stewart, B.W.; Wild, C.P. World Cancer Report; International Agency for Research on Cancer; Lyon, France, 2014, ISBN: 978-92-832-0429-9.
1206 1207
W.H.O. Fact Sheet/February 2018. Available online: http://www.who.int/newsroom/fact-sheets/detail/cancer (accessed on 13th September 2018).
1204 1205
Stratton, M.R.; Campbell, P.J.; Futreal, P.A. The cancer genome. Nature 2009, 458, 719-
5.
Cavaliere,
R.; Ciocatto,
E.C.; Giovanella,
B.C.; Heidelberger,
C.; Johnson,
1210
R.O.; Margottini, M.; Mondovi, B.; Moricca, G.; Rossi-Fanelli, A. Selective heat
1211
sensitivity of cancer cells. Biochemical and clinical studies. Cancer 1967, 20, 1351-81.
1212
6.
Bleehen, N.M. Hyperthermia in the treatment of cancer. Br. J. Cancer. Suppl. 1982, 5, 96-100.
1213 1214
7.
Hofer, K.G. Hyperthermia and cancer. Eur. Cells Mater. 2002, 3, 67-69.
1215
8.
Wust, P.; Hildebrandt, B.; Sreenivasa, G.; Rau, B.; Gellermann, J.; Riess, H.; Felix, R.;
1216
Schlag P.M. Hyperthermia in combined treatment of cancer. Lancet Oncol. 2002, 3, 487-
1217
497.
1218 1219 1220 1221
9.
Jha, S.; Sharma, P.K.; Malviya, R. Hyperthermia: Role and Risk Factor for Cancer Treatment. Achievements in the Life Sciences 2016, 10, 161-167.
10. Melamed, J.R.; Edelstein, R.S.; Day, E.S. Elucidating the Fundamental Mechanisms of Cell Death Triggered by Photothermal Therapy. ACS Nano 2015, 9, 6-11.
56 ACS Paragon Plus Environment
Page 57 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1222
11. Hergt, R.; Dutz, S.; Muller. R.; Zeisberger, M. Magnetic particle hyperthermia:
1223
nanoparticle magnetism and materials development for cancer therapy. J. Phys.:
1224
Condens. Matter 2006, 18, 2919-2934.
1225
12. Zou, L.; Wang, H.; He, B.; Zeng, L.; Tan, T.; Cao, H.; He, X.; Zhang, Z.; Guo, S.; Li, Y.
1226
Current Approaches of Photothermal Therapy in Treating Cancer Metastasis with
1227
Nanotherapeutics. Theranostics 2016, 6, 762–772.
1228 1229 1230 1231 1232 1233
13. Ferrari, M. Cancer nanotechnology: opportunities and challenges. Nature Reviews Cancer 2005, 5, 161-171. 14. Huebsch, N.; Mooney, D.J. Inspiration and application in the evolution of biomaterials. Nature 2009, 462, 426-432. 15. Hench, L.L.; Thompson, I. Twenty-first century challenges for biomaterials. J. R. Soc. Interface 2010, 7, 379-391.
1234
16. Li, F.; Li, T.; Cao, W.; Wang, L.; Xu H., Near-infrared light stimuli-responsive
1235
synergistic therapy nanoplatforms based on the coordination of tellurium-containing
1236
block polymer and cisplatin for cancer treatment. Biomaterials 2017, 133, 208-218.
1237
17. Hoare, T.; Santamaria, J.; Goya, G.F.; Irusta, S.; Lin, D.; Lau, S.; Padera, R.; Langer, R.;
1238
Kohane, D.S. A magnetically triggered composite membrane for on-demand drug
1239
delivery. Nano Lett. 2009, 9, 3651-3657.
1240
18. Zhang, S.; Bellinger, A.M.; Glettig, D.L.; Barman, R.; Lee, Y.-A.L.; Zhu, J.; Cleveland,
1241
C.; Montgomery, V.A.; Gu, L.; Nash, L.D.; Maitland, D.J.; Langer, R.; Traverso, G. A
1242
pH-responsive supramolecular polymer gel as an enteric elastomer for use in gastric
1243
devices. Nature Materials 2015, 14, 1065-1071.
1244
19. Zhernenkov, M.; Ashkar, R.; Feng, H.; Akintewe, O.O.; Gallant, N.D.; Toomey, R.;
1245
Ankner, J.F.; Pynn, R. Thermoresponsive PNIPAM coatings on nanostructured gratings
1246
for cell alignment and release. ACS Appl. Mater. Interfaces, 2015, 7, 11857-11862.
57 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1247 1248
Page 58 of 84
20. Ge, J.; Neofytou, E.; Cahill, T.J.; Beygui, R.E.; Zare, R.N. Drug Release from ElectricField-Responsive Nanoparticles. ACS Nano, 2012, 6, 227-233.
1249 1250
21. Miller, J.G. Living systems: Basic concepts. Behavioral Science 1965, 10, 193-237.
1251
22. Kitano, H. Systems Biology: A Brief Overview. Science 2002, 295, 1662-1664.
1252
23. Grzybowski, B.A.; Huck, W.T.S. The nanotechnology of life-inspired systems. Nature
1253 1254 1255 1256 1257 1258 1259
Nanotechnology 2016, 11, 585-592. 24. Sun, T.; Qing, G. Biomimetic Smart Interface Materials for Biological Applications. Adv. Mater. 2011, 23, 57-77. 25. Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nature Materials 2013, 12, 991-1003. 26. Guragain, S.; Bastakoti, B.P.; Malgras, V.; Nakashima, K.; Yamauchi, Y. Multi-StimuliResponsive Polymeric Materials. Chem. Eur. J. 2015, 21, 13164-13174.
1260
27. Blum, A.P.; Kammeyer, J.K.; Rush, A.M.; Callmann, C.E.; Hahn, M.E.; Gianneschi,
1261
N.C. Stimuli-Responsive Nanomaterials for Biomedical Applications. J. Am. Chem.
1262
Soc. 2015, 137, 2140-2154.
1263 1264
28. Pattani, V.P.; Tunnell, J.W. Nanoparticle-mediated photothermal therapy: A comparative study of heating for different particle types. Lasers Surg. Med. 2012, 44, 675-684.
1265
29. Jain, P.K.; Huang, X.; El-Sayed, I.H.; El-Sayed, M.A. Review of Some Interesting
1266
Surface Plasmon Resonance-enhanced Properties of Noble Metal Nanoparticles and Their
1267
Applications to Biosystems. Plasmonics 2007, 2, 107-118.
1268
30. De Sio, L.; Placido, T.; Comparelli, R.; Lucia Curri, M.; Striccoli, M.; Tabiryan, N.;
1269
Bunning,
T.J.
Next-generation
thermo-plasmonic
1270
nanoparticles in optoelectronics. Prog. Quantum Electron. 2015, 41, 23-70.
58 ACS Paragon Plus Environment
technologies
and
plasmonic
Page 59 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1271
31. Gunduz, N.; Ceylan, H.; Guler, M.O.; Tekinay, A.B. Intracellular Accumulation of Gold
1272
Nanoparticles Leads to Inhibition of Macropinocytosis to Reduce the Endoplasmic
1273
Reticulum Stress. Scientific Reports 2017, 7, 40493.
1274 1275
32. Markovic, Z.M.; Harhaji-Trajkovic, L.M.; Todorovic-Markovic, B.M.; Kepić, D.P.;
1276
Arsikin, K.M.; Jovanović, S.P.; Pantovic, A.C.; Dramićanin, M.D.; Trajkovic, V.S. In
1277
vitro comparison of the photothermal anticancer activity of graphene nanoparticles and
1278
carbon nanotubes. Biomaterials 2011, 32, 1121-1129.
1279 1280
33. Yang, K.; Li, Y.; Tan, X.; Peng, R.; Liu, Z. Behavior and Toxicity of Graphene and Its Functionalized Derivatives in Biological Systems. Small 2013, 9, 1492-1503.
1281
34. Gualandi, C.; Zucchelli, A.; Fernández Osorio, M.; Belcari, J.; Focarete, M.L.
1282
Nanovascularization of polymer matrix: generation of nanochannels and nanotubes by
1283
sacrificial electrospun fibers. Nano Lett. 2013, 13, 5385-5390.
1284
35. Nakielski, P.; Pawłowska, S.; Pierini, F.; Liwińska, W.; Hejduk, P.; Zembrzycki, K.;
1285
Zabost, E.; Kowalewski, T.A. Hydrogel Nanofilaments via Core-Shell Electrospinning.
1286
PLoS ONE 2015, 10, 0129816.
1287
36. Sun, K.; Yang, Y.; Zhou, H.; Yin, S.; Qin, W.; Yu, J.; Chiu, D.T.; Yuan, Z.; Zhang,
1288
X.; Wu, C. Ultrabright Polymer-Dot Transducer Enabled Wireless Glucose Monitoring
1289
via a Smartphone. ACS Nano 2018, 12, 5176-5184.
1290
37. Preem, L.; Mahmoudzadeh, M.; Putrinš, M,; Meos, A.; Laidmäe, I.; Romann, T.; Aruväli,
1291
J.; Härmas, R.; Koivuniemi, A.; Bunker, A.; Tenson, T.; Kogermann, K. Interactions
1292
between Chloramphenicol, Carrier Polymers, and Bacteria–Implications for Designing
1293
Electrospun Drug Delivery Systems Countering Wound Infection. Mol. Pharmaceutics
1294
2017, 14, 4417-4430.
59 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1295
38. Stachewicz, U.; Qiao, T.; Rawlinson, S.C.F.; Almeida, F.V.; Li, W.-Q.; Cattell, M.;
1296
Barber, A.H. 3D imaging of cell interactions with electrospun PLGA nanofiber
1297
membranes for bone regeneration. Acta Biomaterialia 2015, 27, 88-100.
1298 1299 1300 1301
39. Xu, L.; Cheng, L.; Wang, C.; Peng, R.; Liu, Z. Conjugated polymers for photothermal therapy of cancer. Polym. Chem. 2014, 5, 1573-1580. 40. Mrowczynski, R. Polydopamine-Based Multifunctional (Nano)materials for Cancer Therapy. ACS Appl. Mater. Interfaces 2018, 10, 7541-7561.
1302
41. Geng, J.; Sun, C.; Liu, J.; Liao, L.-D.; Yuan, Y.; Thakor, N.; Wang, J.; Liu B.
1303
Biocompatible Conjugated Polymer Nanoparticles for Efficient Photothermal Tumor
1304
Therapy. Small 2015, 11, 1603-1610.
1305 1306
42. Kobayashi, S. Green polymer chemistry: new methods of polymer synthesis using renewable starting materials. Struct. Chem. 2017 28, 461-474.
1307
43. Rinoldi, C.; Kijenska, E.; Chlanda, A.; Choinska, E.; Khenoussi, N.; Tamayol, A.;
1308
Khademhosseini, A.; Swieszkowski, W. Nanobead-on-string composites for tendon tissue
1309
engineering. J. Mater. Chem. B, 2018, 6, 3116-3127.
1310
44. Costantini, M.; Testa, S.; Mozetic, P.; Barbetta, A.; Fuoco, C.; Fornetti, E.; Tamiro, F.;
1311
Bernardini, S.; Jaroszewicz, J.; Swieszkowski, W.; Trombetta, M.; Castagnoli, L.;
1312
Seliktar, D.; Garstecki, P.; Cesareni, G.; Cannata, S.; Rainer, A.; Gargioli, C.
1313
Microfluidic-enhanced 3D bioprinting of aligned myoblast-laden hydrogels leads to
1314
functionally organized myofibers in vitro and in vivo. Biomaterials 2017, 131, 98-110.
1315
45. Ha, W.; Zhao, X.B.; Jiang, K.; Kang, Y.; Chen, J.; Li, B. J.; Shi, Y. P. A three-
1316
dimensional graphene oxide supramolecular hydrogel for infrared light-responsive
1317
cascade release of two anticancer drugs. Chem. Commun. 2016, 52, 14384-14387.
1318 1319
46. Zhou, Y.; Ye, H.; Chen, Y.; Zhu, R.; Yin, L. Photoresponsive Drug/Gene Delivery Systems. Biomacromolecules 2018, 19, 1840-1857.
60 ACS Paragon Plus Environment
Page 60 of 84
Page 61 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1320
47. Liang, Y.; Chen, Z.; Jing, Y.; Rong, Y.; Facchetti, A.; Yao, Y. Heavily n-dopable π-
1321
donjugated redox polymers with ultrafast energy storage capability. J. Am. Chem. Soc.
1322
2015, 137, 4956-4959.
1323 1324
48. Pierini, F.; Lanzi, M.; Nakielski, P.; Pawłowska, S.; Zembrzycki, K.; Kowalewski, T.A.
1325
Electrospun poly (3‐hexylthiophene)/poly (ethylene oxide)/graphene oxide composite
1326
nanofibers: effects of graphene oxide reduction. Polym. Adv. Technol. 2016, 27, 1465-
1327
1475.
1328
49. Pierini, F.; Lanzi, M.; Nakielski, P.; Pawłowska, S.; Urbanek, O.; Zembrzycki, K.;
1329
Kowalewski, T.A. Single-material organic solar cells based on electrospun fullerene-
1330
grafted polythiophene nanofibers. Macromolecules 2017, 50, 4972-4981.
1331
50. Chen, X.; Li, R.; Liu, Z.; Sun, K.; Sun, Z.; Chen, D.; Xu, G.; Xi, P.; Wu, C.; Sun, Y.
1332
Small Photoblinking Semiconductor Polymer Dots for Fluorescence Nanoscopy. Adv.
1333
Mater. 2017, 29, 1604850.
1334
51. Molina, M.; Wedepohl, S.; Calderón, M. Polymeric near-infrared absorbing dendritic
1335
nanogels for efficient in vivo photothermal cancer therapy. Nanoscale 2016, 8, 5852-
1336
5856.
1337
52. Wang, J.; Yan, R.; Guo, F.; Yu, M.; Tan, F.; Li, N. Targeted lipid–polyaniline hybrid
1338
nanoparticles for photoacoustic imaging guided photothermal therapy of cancer.
1339
Nanotechnology 2016, 27, 285102.
1340
53. Bharathiraja, S.; Manivasagan, P.; Oh, Y.O.; Moorthy, M. S.; Seo, H.; Bui, N. Q.; Oh, J.
1341
Astaxanthin conjugated polypyrrole nanoparticles as a multimodal agent for photo-based
1342
therapy and imaging. Int. J. Pharm. 2017, 517, 216-225.
61 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 62 of 84
1343
54. Wang, X.; Ma, Y.; Sheng, X.; Wang, Y.; Xu, H. Ultrathin polypyrrole nanosheets via
1344
space-confined synthesis for efficient photothermal therapy in the second near-infrared
1345
window. Nano Lett. 2018, 18, 2217-2225.
1346 1347 1348
55. Ren, S.; Cheng, X.; Chen, M.; Liu, C.; Zhao, P.; Huang, W.; He, J.; Zhou, Z.; Miao, L.
1349
Hypotoxic and rapidly metabolic PEG-PCL-C3-ICG nanoparticles for fluorescence-
1350
guided photothermal/photodynamic therapy against OSCC. ACS Appl. Mater. Interfaces
1351
2017, 9, 31509-31518.
1352
56. Lyu, Y.; Zeng, J.; Jiang, Y.; Zhen, X.; Wang, T.; Qiu, S.; Lou, X.; Gao, M.; Pu, K.
1353
Enhancing
Both
Biodegradability
and
Efficacy
of
Semiconducting
Polymer
1354
Nanoparticles for Photoacoustic Imaging and Photothermal Therapy. ACS Nano 2018, 12,
1355
1801-1810.
1356
57. Wang, X.H.; Peng, H.S.; Yang, W.; Ren, Z.D.; Liu, X.M.; Liu, Y.A. Indocyanine green-
1357
platinum porphyrins integrated conjugated polymer hybrid nanoparticles for near-
1358
infrared-triggered photothermal and two-photon photodynamic therapy. J. Mater. Sci. B.
1359
2017, 5, 1856-1862.
1360
58. Cao, Z.; Feng, L.; Zhang G.; Wang, J.; Shen S.; Li, D.; Yang X. Semiconducting
1361
polymer-based nanoparticles with strong absorbance in NIR-II window for in vivo
1362
photothermal therapy and photoacoustic imaging. Biomaterials 2018, 155, 103-111.
1363 1364
59. Jiang, Y.; Cui, D.; Fang, Y.; Zhen, X.; Uppunturi, P.K.; Pramanik, M.; Ding, D.; Pu, K. Amphiphilic
semiconducting
polymer
as
multifunctional
62 ACS Paragon Plus Environment
nanocarrier
for
Page 63 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1365
fluorescence/photoacoustic imaging guided chemo-photothermal therapy. Biomaterials
1366
2017, 145, 168-177.
1367
60. Yuan, Y.; Wang, Z.; Cai, P.; Liu, J.; Liao, L.D.; Hong, M.; Chen, X.; Thakor, N.; Liu, B.
1368
Conjugated polymer and drug co-encapsulated nanoparticles for chemo- and photo-
1369
thermal combination therapy with two-photon regulated fast drug release. Nanoscale
1370
2015, 7, 3067-3076.
1371
61. Feng, G.; Fang, Y.; Liu, J.; Geng, J.; Ding, D.; Liu, B. Multifunctional conjugated
1372
polymer nanoparticles for image-guided photodynamic and photothermal therapy. Small
1373
2017, 13, 1602807.
1374
62. Guo, B.; Feng, G.; Manghnani, P.N.; Cai, X.; Liu, J.; Wu, W.; Xu, S.; Cheng, X.; Teh,
1375
C.; Liu, Bin. A porphyrin-based conjugated polymer for highly efficient in vitro and in
1376
vivo photothermal therapy. Small 2016, 12, 6243-6254.
1377
63. Cai, X.; Liu, X.; Liao, L.D.; Bandla, A.; Ling, J.M.; Liu, Y.H.; Thakor, N.; Bazan, G.C.;
1378
Liu, B. Encapsulated conjugated oligomer nanoparticles for real-time photoacoustic
1379
sentinel lymph node imaging and targeted photothermal therapy. Small 2016, 12, 4873-
1380
4880.
1381
64. Cao, Y.; Dou, J.H., Zhao, N.J.; Zhang, S.; Zheng, Y.Q; Zhang, J.P.; Wang, J.Y.; Pei, J.;
1382
Wang, Y.
Highly efficient NIR-II photothermal conversion based on an organic
1383
conjugated polymer. Chem. Mater. 2017, 29, 718-725.
1384
65. Huang, S.; Upputuri, P.K.; Liu, H.; Pramanik, M.; Wang, M. A dual-functional
1385
benzobisthiadiazole derivative as an effective theranostic agent for near-infrared
1386
photoacoustic imaging and photothermal therapy. J. Mater. Chem. B. 2016, 4, 1696-1703.
63 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1387
66. Li, D.-D.; Wang, J.X.; Ma, Y.; Qian, H.-S.; Wang, D.; Wang, L.; Zhang, G.; Qiu, L.;
1388
Wang, Y.-C.; Yang, X.-Z. A donor−acceptor conjugated polymer with alternating
1389
isoindigo derivative and bithiophene units for near-infrared modulated cancer thermo-
1390
chemotherapy. ACS Appl. Mater. Interfaces 2016, 8, 19312-19320.
1391
67. Zhang, J.; Yang, C.; Zhang, R.; Chen, R.; Zhang, Z.; Zhang, W.; Peng, S.H.; Chen, X.;
1392
Liu, G.; Hsu, C.S.; Lee, C.S. Biocompatible D–A semiconducting polymer nanoparticle
1393
with light-harvesting unit for highly effective photoacoustic imaging guided photothermal
1394
therapy. Adv. Funct. Mater. 2017, 27, 1605094.
1395
68. Chang, K.; Liu, Y.; Hu, D.; Qi, Q.; Gao, D.; Wang, Y.; Li, D.; Zhang, X.; Zheng, H.;
1396
Sheng, Z.; Yuan, Z. Highly Stable Conjugated Polymer Dots as Multifunctional Agents
1397
for Photoacoustic Imaging-Guided Photothermal Therapy. ACS Appl. Mater. Interfaces
1398
2018 10, 7012-7021.
1399
69. Chen, H.; Zhang, J.; Chang, K.; Men, X.; Fang, X.; Zhou, L.; Li, D.; Gao, D.; Yin, S.;
1400
Zhang, X.; Yuan, Z.; Wu, C. Highly absorbing multispectral near-infrared polymer
1401
nanoparticles from one conjugated backbone for photoacoustic imaging and photothermal
1402
therapy. Biomaterials 2017, 144, 42-52.
1403
70. Chang, K.; Liu, Z.; Fang, X.; Chen, H.; Men, X.; Yuan, Y.; Sun, K.; Zhang, X.; Yuan, Z.;
1404
Wu, C. Enhanced Phototherapy by Nanoparticle-Enzyme via Generation and Photolysis
1405
of Hydrogen Peroxide; Nano Lett. 2017, 17, 4323-4329.
1406
71. Wu, C.; Hansen, S.J.; Hou, Q.; Yu, J.; Ziegler, M.; Jin, Y.; Burnham, D.R.; McNeill,
1407
J.D.; Olson, J.M.; Chiu, D.T. Design of Highly Emissive Polymer Dot Bioconjugates for
1408
In Vivo Tumor Targeting. Angew. Chem. 2011, 123, 3492-3496.
64 ACS Paragon Plus Environment
Page 64 of 84
Page 65 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1409
72. Chen, D.; Wu, I.-C.; Liu, Z.; Tang, Y.; Chen, H.; Yu, J.; Wu, C.; Chiu, D.T.
1410
Semiconducting polymer dots with bright narrow-band emission at 800 nm for biological
1411
applications. Chem. Sci. 2017, 8, 3390-3398.
1412 1413 1414 1415
73. Jia, H.; Titmuss, S. Polymer-functionalized nanoparticles: from stealth viruses to biocompatible quantum dots. Nanomedicine 2009, 4, 951–996. 74. Li, D.; He, Q.; Li, J. Smart core/shell nanocomposites: Intelligent polymers modified gold nanoparticles. Advances in Colloid and Interface Science 2009, 149, 28–38.
1416
75. Yang, S.-T.; Shiral Fernando, K. A.; Liu, J.-H.; Wang, J.; Sun, H.-F.; Liu, Y.; Chen, M.;
1417
Huang, Y.; Wang, X.; Wang, H.; Sun, Y.-P. Covalently PEGylated Carbon Nanotubes
1418
with Stealth Character In Vivo. Small 2008, 4, 940–944.
1419
76. Chen, H.; Zhou, H.; Men, X.; Sun, K.; Sun, Z.; Fang, X.; Wu, Ch. Light-Induced
1420
PEGylation and Functionalization of Semiconductor Polymer Dots. Chem. Nano. Mat.
1421
2017, 3, 755-759.
1422 1423
77. Li, K.-C.; Chu, H.-C.; Lin, Y.; Tuan, H.-Y.; Hu, Y.-C. PEGylated Copper Nanowires as a Novel Photothermal Therapy Agent. ACS Appl. Mater. Interfaces 2016, 8, 12082–12090.
1424
78. O’Neal, D.P.; Hirsch, L.R.; Halas, N.J.; Payne, J.D.; West, J.L. Photo-thermal tumor
1425
ablation in mice using near infrared-absorbing nanoparticles. Cancer Letters 2004, 209,
1426
171–176.
1427
79. Hirsch, L.R.; Staffordt, R.J.; Banksont, J.A.; Sershen, S.R.; B. Rivera, Prices, R. E.;
1428
Hazlet, J. D.; Halass, N.J.; West, J.L. Nanoshell-mediated near-infrared thermal therapy
1429
of tumors under magnetic resonance guidance. PNAS 2003, 11, 13549-13554.
65 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1430
80. Yang, K.; Zhang, Sh.; Zhang, G.; Sun, X.; Lee, Sh.-T.; Liu, Z. Graphene in Mice:
1431
Ultrahigh In Vivo Tumor Uptake and Efficient Photothermal Therapy. Nano Lett. 2010,
1432
10, 3318–3323.
1433
81. Lin, W.; Li, Y.; Zhang, W.; Liu, S.; Xie, Z.; Jing, X. Near-infrared polymeric
1434
nanoparticles with high content of cyanine for bimodal imaging and photothermal
1435
therapy. ACS Appl. Mater. Interfaces 2016, 8, 24426-24432.
1436
82. Boca, S.C.; Potara, M.; Gabudean, A.-M.; Juhem, A.; Baldeck, P.L.; Astilean, S.
1437
Chitosan-coated triangular silver nanoparticles as a novel class of biocompatible, highly
1438
effective photothermal transducers for in vitro cancer cell therapy. Cancer Letters 2011,
1439
311, 131–140.
1440
83. Salem, D.S.; Sliem, M.A.; El-Sesy, M.; Shouman, S.A.; Badr, Y. Improved chemo-
1441
photothermal therapy of hepatocellular carcinoma using chitosan-coated gold
1442
nanoparticles. Journal of Photochemistry & Photobiology, B: Biology 2018, 182, 92–99.
1443
84. Zhang, G.; Sun, X.; Jasinski, J.; Patel, D.; Gobin, A. M. Gold/Chitosan Nanocomposites
1444
with Specific Near Infrared Absorption for Photothermal Therapy Applications. Journal
1445
of Nanomaterials 2012, 2012, 1-9.
1446
85. Daniels, J.L.; Crawford, T.M.; Andreev, O.A.; Reshetnyak, Y.K. Synthesis and
1447
characterization of pHLIP coated gold nanoparticles. Biochemistry and Biophysics
1448
Reports 2017, 10, 62–69.
1449
86. Otari, S.V.; Kumar, M.; Zahid Anwar, M.; Thorat, N.D.; Patel, S.K.S.; Lee, D.; Lee, J.H.;
1450
Lee, J.-K.; Chan Kang, J.; Zhang, L. Rapid synthesis and decoration of reduced graphene
1451
oxide with gold nanoparticles by thermostable peptides for memory device and
1452
photothermal applications. Scientific Reports 2017, 7, 10980.
66 ACS Paragon Plus Environment
Page 66 of 84
Page 67 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1453
87. Tan, H.; Huang, Y.; Xu, J.; Chen, B.; Zhang, P.; Ye, Z.; Liang, S.; Xiao, L.; Liu, Z.
1454
Spider Toxin Peptide Lycosin-I Functionalized Gold Nanoparticles for in vivoTumor
1455
Targeting and Therapy. Theranostics 2017, 7, 3168-3178.
1456
88. Shangguan, D.; Li, Y.; Tang, Z.; Cao, Z. Ch.; Chen, H.W.; Mallikaratchy, P.; Sefah, K.;
1457
Yang, Ch. J.; Tan W. Aptamers evolved from live cells as effective molecular probes for
1458
cancer study. PNAS 2006, 32, 11838-11843.
1459
89. Riley, R.S.; Day, E.S. Gold nanoparticle-mediated photothermal therapy: applications
1460
and opportunities for multimodal cancer treatment. WIREs Nanomed. Nanobiotechnol.
1461
2017, 9, 1449.
1462
90. Wang, B.-K.; Yu, X.-F.; Wang, J.-H.; Li, Z.-B.; Li, P.-H.; Wang, H.; Song, L.; Chu, P.
1463
K.; Li, Ch. Gold-nanorods-siRNA nanoplex for improved photothermal therapy by gene
1464
silencing. Biomaterials 2016, 78, 27-39.
1465
91. Braun, G.B.; Pallaoro, A.; Wu, G.; Missirlis, D.; Zasadzinski, J. A.; Tirrell, M.; Reich, N.
1466
O. Laser-activated gene silencing via gold nanoshell-siRNA conjugates. ACS Nano 2009,
1467
3, 2007–2015.
1468 1469
92. Takahashi, H.; Niidome, Y.; Yamada, S. Controlled release of plasmid DNA from gold nanorods induced by pulsed near-infrared light. Chem. Commun. 2005, 2247–2249.
1470
93. Ocsoy, I.; Yusufbeyoglu, S.; Yılmaz, V.; McLamore, E. S.; Ildız, N.; Ülgen, A. DNA
1471
aptamer functionalized gold nanostructures for molecular recognition and photothermal
1472
inactivation of methicillin-Resistant Staphylococcus aureus. Colloids and Surfaces B:
1473
Biointerfaces 2017, 159, 16–22.
1474 1475
94. Shalin, R.E. In Polymer matrix composites, Vol. 4; Springer Science & Business Media: London, UK, 2012.
67 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 68 of 84
1476
95. Valerio, M.; Ahmed, H.U.; Emberton, M.; Lawrentschuk, N.; Lazzeri, M.; Montironi, R.;
1477
Nguyen P.L.; Trachtenberg J.; Polascik, T. J. The role of focal therapy in the
1478
management of localised prostate cancer: a systematic review. Eur. Urol. 2014, 66 (4),
1479
732-751.
1480 1481
96. Sill, T.J.; von Recum, H.A. Electrospinning: applications in drug delivery and tissue engineering. Biomaterials 2008, 29 (13), 1989-2006.
1482
97. Sosnik, A.; das Neves, J.; Sarmento, B. Mucoadhesive polymers in the design of nano-
1483
drug delivery systems for administration by non-parenteral routes: a review. Prog. Polym.
1484
Sci. 2014, 39(12), 2030-2075.
1485 1486
98. Damia, G.; Garattini, S. The pharmacological point of view of resistance to therapy in tumors. Cancer Treat. Rev. 2014, 40 (8), 909-916.
1487
99. Conde, J.; Oliva, N.; Zhang, Y.; Artzi, N. Local triple-combination therapy results in
1488
tumour regression and prevents recurrence in a colon cancer model. Nat. Mater.
1489
2016, 15(10), 1128.
1490
100.Zhao, P.; Zheng, M.; Yue, C.; Luo, Z.; Gong, P.; Gao, G.; Sheng, Z.; Zheng, C.; Cai, L.
1491
Improving drug accumulation and photothermal efficacy in tumor depending on size of
1492
ICG loaded lipid-polymer nanoparticles. Biomaterials 2014, 35 (23), 6037-6046.
1493 1494 1495
101.Petros, R.A.; DeSimone, J.M. Strategies in the design of nanoparticles for therapeutic applications. Nat. Rev. Drug. Discov. 2010, 9(8), 615. 102.Zhang, Z.; Liu, S.; Xiong, H.; Jing, X.; Xie, Z.; Chen, X.; Huang, Y. Electrospun
1496
PLA/MWCNTs
composite
nanofibers
1497
therapy. Acta Biomater. 2015, 26, 115-123.
for
combined
68 ACS Paragon Plus Environment
chemo-and
photothermal
Page 69 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1498
103.Viswanath, V.; Maity, S.; Bochinski, J.R.; Clarke, L. I.; Gorga, R. E. Thermal annealing
1499
of polymer nanocomposites via photothermal heating: effects on crystallinity and
1500
spherulite morphology. Macromolecules 2013, 46 (21), 8596-8607.
1501 1502 1503 1504
104.You, Y.; Lee, S.W.; Lee, S.J.; Park, W.H. Thermal interfiber bonding of electrospun poly (l-lactic acid) nanofibers. Mater. Lett. 2006, 60 (11), 1331-1333. 105.Zia, Q.; Ingolič, E.; Androsch, R. Surface and bulk morphology of cold-crystallized poly (ethylene terephthalate). Colloid Polym. Sci. 2010, 288 (7), 819-825.
1505
106.Estrada, A. C.; Daniel-da-Silva, A.L.; Trindade, T. Photothermally enhanced drug release
1506
by κ-carrageenan hydrogels reinforced with multi-walled carbon nanotubes. RSC Adv.
1507
2013, 3 (27), 10828-10836.
1508 1509
107.Caló, E.; Khutoryanskiy, V.V. Biomedical applications of hydrogels: A review of patents and commercial products. Eur. Polym. J. 2015, 65, 252-267.
1510
108.Yoo, M.K..; Sung, Y. K.; Lee, Y.M.; Cho, C. S. Effect of polyelectrolyte on the lower
1511
critical solution temperature of poly (N-isopropyl acrylamide) in the poly (NIPAAm-co-
1512
acrylic acid) hydrogel. Polymer 2000, 41 (15), 5713-5719.
1513 1514
109.Ruel-Gariepy, E.; Leroux, J.C. In situ-forming hydrogels—review of temperaturesensitive systems. Eur. J. Pharm. Biopharm. 2004, 58 (2), 409-426.
1515
110.Xing, R.; Liu, K..; Jiao, T.; Zhang, N.; Ma, K.; Zhang, R.; Zou Q.; Ma G.; Yan, X. An
1516
Injectable Self‐Assembling Collagen–Gold Hybrid Hydrogel for Combinatorial
1517
Antitumor Photothermal/Photodynamic Therapy. Adv. Mater. 2016, 28 (19), 3669-3676.
1518 1519
111.Chang, G.; Li, S.; Huang, F.; Zhang, X.; Shen, Y.; Xie, A. Multifunctional reduced graphene
oxide
hydrogel
as
drug
carrier
for
69 ACS Paragon Plus Environment
localized
and
synergic
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1520
photothermal/photodynamics/chemo therapy. J. Mater. Sci. Technol. 2016, 32(8), 753-
1521
762.
1522
112.Wu, W.; Shen, J.; Banerjee, P.; Zhou, S.; Core–shell hybrid nanogels for integration of
1523
optical temperature-sensing, targeted tumor cell imaging, and combined chemo-
1524
photothermal treatment. Biomaterials 2010, 31 (29), 7555-7566.
1525
113.Zhang, H.; Zhu, X.; Ji, Y.; Jiao, X.; Chen, Q.; Hou, L.; Zhang, H.; Zhang, Z. Near-
1526
infrared-triggered in situ hybrid hydrogel system for synergistic cancer therapy. J. Mater.
1527
Chem. B 2015, 3 (30), 6310-6326.
1528
114.Meinhardt, M.; Krebs, R.; Anders, A.; Heinrich, U.; Tronnier, H. Wavelength-dependent
1529
penetration depths of ultraviolet radiation in human skin. J. Biomed. Opt. 2008, 13 (4),
1530
044030.
1531
115.Stolik, S.; Delgado, J. A.; Perez, A.; Anasagasti, L. Measurement of the penetration
1532
depths of red and near infrared light in human “ex vivo” tissues. J. Photoch. Photobio. B
1533
2000, 57 (2), 90-93.
1534
116.Zhou, M.; Liu, S.; Jiang, Y.; Ma, H.; Shi, M.; Wang, Q.; Zhong W.; Liao W.; Xing, M.
1535
M. Doxorubicin‐Loaded Single Wall Nanotube Thermo‐Sensitive Hydrogel for Gastric
1536
Cancer Chemo‐Photothermal Therapy. Adv. Funct. Mater. 2015, 25 (29), 4730-4739.
1537
117.Schlagenhauf, L.; Buerki-Thurnherr, T.; Kuo, Y.Y.; Wichser, A.; Nüesch, F.; Wick, P.;
1538
Wang, J. Carbon nanotubes released from an epoxy-based nanocomposite: quantification
1539
and particle toxicity. Environ. Sci. Technol. 2015, 49 (17), 10616-10623.
1540
118.Zhang, Y.; Ali, S. F.; Dervishi, E.; Xu, Y.; Li, Z.; Casciano, D.; Biris, A. S. Cytotoxicity
1541
effects of graphene and single-wall carbon nanotubes in neural phaeochromocytoma-
1542
derived PC12 cells. ACS Nano 2010, 4 (6), 3181-3186.
70 ACS Paragon Plus Environment
Page 70 of 84
Page 71 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1543 1544
Biomacromolecules
119.Brown, T.D.; Dalton, P.D.; Hutmacher, D.W. Melt electrospinning today: An opportune time for an emerging polymer process. Prog. Polym. Sci. 2016, 56, 116-166.
1545
120.Dulnik, J., Kołbuk, D., Denis, P., Sajkiewicz, P. The effect of a solvent on cellular
1546
response to PCL/gelatin and PCL/collagen electrospun nanofibres. Eur.Polym. J.
1547
2018, 104, 147-156.
1548
121.Urbanek, O., Sajkiewicz, P., Pierini, F., Czerkies, M., Kołbuk, D. Structure and
1549
properties of polycaprolactone/chitosan nonwovens tailored by solvent systems. Biomed.
1550
Mater. 2017, 12 (1), 015020.
1551
122.Cheng, M.; Wang, H.; Zhang, Z.; Li, N.; Fang, X.; Xu, S. Gold nanorod-embedded
1552
electrospun fibrous membrane as a photothermal therapy platform. ACS Appl. Mater.
1553
Inter. 2014, 6 (3), 1569-1575.
1554 1555
123.Greiner, A.; Wendorff, J.H.; Yarin, A. L.; Zussman E. Biohybrid nanosystems with polymer nanofibers and nanotubes. Appl. Microbiol. Biotechnol. 2006, 71, 387-393.
1556
124.Yu, Q.; Han, Y.; Wang, X.; Qin, C.; Zhai, D.; Yi, Z.; Chang J.; Xiao Y.; Wu, C. Copper
1557
Silicate Hollow Microspheres-Incorporated Scaffolds for Chemo-Photothermal Therapy
1558
of Melanoma and Tissue Healing. ACS Nano 2018, 12, 2695-2707.
1559
125.Lin, W.; Li, Q.; Zhu, T. Study of solvent casting/particulate leaching technique
1560
membranes in pervaporation for dehydration of caprolactam. J Ind Eng Chem 2012, 18
1561
(3), 941-947.
1562
126.Campbell, J. J.; Husmann, A.; Hume, R. D.; Watson, C. J.; Cameron, R. E. Development
1563
of three-dimensional collagen scaffolds with controlled architecture for cell migration
1564
studies using breast cancer cell lines. Biomaterials 2017, 114, 34-43.
71 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1565
127.Shao, J.; Xie, H.; Wang, H.; Zhou, W.; Luo, Q.; Yu, X. F.; Chu, P. K. 2D material-based
1566
nanofibrous membrane for photothermal cancer therapy. ACS Appl. Mater. & Interfaces
1567
2017, 10 (1), 1155-1163.
1568
128.Lee, J. H.; Park, T. G.; Park, H. S.; Lee, D. S.; Lee, Y. K.; Yoon, S. C.; Nam, J. D.
1569
Thermal and mechanical characteristics of poly (L-lactic acid) nanocomposite
1570
scaffold. Biomaterials 2003, 24 (16), 2773-2778.
1571
129.Yoo, H.S.; Kim, T.G.; Park, T.G. Surface-functionalized electrospun nanofibers for
1572
tissue engineering and drug delivery. Adv. Drug. Delivery Rev. 2009, 61 (12), 1033-1042.
1573
130.Li, Y.; Verbiest, T.; Vankelecom, I. Improving the flux of PDMS membranes via
1574
localized heating through incorporation of gold nanoparticles. J. Memb. Sci. 2013, 428,
1575
63-69.
1576
131.Vanherck, K..; Hermans, S.; Verbiest, T.; Vankelecom, I. Using the photothermal effect
1577
to improve membrane separations via localized heating. J. Mater. Chem. A 2011, 21 (16),
1578
6079-6087.
1579
132.Vanherck, K.; Vankelecom, I.; Verbiest, T. Improving fluxes of polyimide membranes
1580
containing gold nanoparticles by photothermal heating. J. Membrane Sci. 2011, 373 (1),
1581
5-13.
1582
133.Wang, C.; Xu, L.; Liang, C.; Xiang, J.; Peng, R.; Liu, Z. Immunological Responses
1583
Triggered by Photothermal Therapy with Carbon Nanotubes in Combination with Anti‐
1584
CTLA‐4 Therapy to Inhibit Cancer Metastasis. Adv. Mater. 2014, 26, 8154-8162.
1585
134.Kunjachan, S.; Detappe, A.; Kumar, R.; Ireland, T.; Cameron, L.; Biancur, D.E.; Motto-
1586
Ros, V.; Sancey, L.; Sridhar, S.; Makrigiorgos, G.M.; Berbeco, R.I. Nanoparticle
72 ACS Paragon Plus Environment
Page 72 of 84
Page 73 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1587
Mediated Tumor Vascular Disruption: A Novel Strategy in Radiation Therapy. Nano
1588
Lett. 2015, 15 (11), 7488-7496.
1589
135.Fu, Y.; Liu, H.; Ren, Z.; Li, X.; Huang, J.; Bestc, S.; Han, G. Luminescent CaTiO3:Yb,Er
1590
nanofibers co-conjugated with Rose Bengal and gold nanorods for potential synergistic
1591
photodynamic/photothermal therapy. J. Mater. Chem. B 2017, 5, 5128-5136.
1592
136.Park, J.H.; Choi, J. H.; Son, J.H.; Hwang S.J.; Seo H.; Kang, I-K.; Park, M.; Kim, J.;
1593
Hyun, D.C. Poly(ε-caprolactone) (PCL) fibers incorporated with phase-changeable fatty
1594
acid and indocyanine green for NIR light-triggered, localized anti-cancer drug release.
1595
Polymer 2018, 135, 211-218.
1596
137.Liu, H.; Fu, Y.; Li, Y.; Ren, Z.; Li, X.; Han, G.; Mao, C. A Fibrous Localized Drug
1597
Delivery Platform with NIR-Triggered and Optically Monitored Drug Release. Langmuir
1598
2016, 32 (35), 9083-9090.
1599
138.Mauro, N.; Scialabba, C.; Pitarresi, G.; Giammona, G. Enhanced adhesion and in situ
1600
photothermal ablation of cancer cells in surface-functionalized electrospun microfiber
1601
scaffold with graphene oxide, Int. J. Pharm. 2017, 526, 167-177.
73 ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 1. (a) Scheme illustrating the biochemical mechanism resulting from nanomaterial-mediated photothermal therapy. Reprinted with permission from ref. 10. Copyright 2015 American Chemical Society. (b) The number of published articles on polymer-based nanomaterials for photothermal therapy during the period 2006–2017, obtained from Web of Science database. 178x60mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 74 of 84
Page 75 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 2. (a) Schematic illustration of the preparation of PPy nanosheets. (b) Fluorescent images at different time points post intravenous injection of PPy nanosheets. (c) Infrared thermographic images of tumorbearing mice after intravenous injection of PBS and PPy nanosheets after irradiation by a 1064 nm laser. Reprinted with permission from ref. 54. Copyright 2018 American Chemical Society. 178x119mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 3. (a) Synthetic route of C3. Reprinted with permission from ref. 55. Copyright 2017 American Chemical Society. (b) Synthesis of DPPV ((i) Pd2(dba)3 and tri(o-tolyl)phosphine, toluene, 100°C, 24 h) and schematic illustration of DPPV/PLGA-PEG NPs. Reprinted with permission from ref. 56. Copyright 2018 American Chemical Society. 178x75mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 76 of 84
Page 77 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 4. Schematic illustration of multifunctional ICG-PtTFPP NPs. Republished with permission of Royal Society of Chemistry, from ref. 57, Copyright 2017; permission conveyed through Copyright Clearance Center, Inc. 178x80mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 5. (a) Schematic illustration of PBTPBF-BT NPs. Reprinted from ref. 58. Copyright 2017, with permission from Elsevier. (b) Chemical structure of TBDOPV-DT polymer. Reprinted with permission from ref. 64. Copyright 2016 American Chemical Society. 178x50mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 78 of 84
Page 79 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 6. (a) Scheme representing the use of PEGylated copper nanowires as a photothermal agent for cancer therapy. (b) Tumor volume of CT26-bearing BALB/c mice after PEGylated copper nanowires after photothermal ablation. Reprinted with permission from ref. 77. Copyright 2016 American Chemical Society. 178x65mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 7. (a) Tumor growth percentage from different groups during the first 18 days after Gold nanorodsiRNA treatment. (b) Photographs of tumors 18 days after G1-G5 treatments. Reprinted from ref. 90. Copyright 2015, with permission from Elsevier. 178x75mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 80 of 84
Page 81 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 8. Images of poly(ethylene oxide) (PEO) spherulites observed through a cross-polarized optical microscope of: (a) melt-crystallized gold nanoparticle:PEO film; (b, c, d) after conventional annealing at 60 °C and after photothermal process; (e, f, g) at different time points; (h) crystallinity; (i) spherulite density as a function of annealing time for gold nanoparticle:PEO melt-crystallized films. The annealing was conducted by conventional and photothermal heating. Reprinted with permission from ref. 103. Copyright 2013 American Chemical Society. 177x113mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 9. (a) Schematic of the smart hydrogel–nanoparticle patch with drug–siRNA nanoparticle conjugates (drug–gold nanorods and siRNA–gold nanospheres) for local gene and drug release designed by Conde et al.99. (b) Fluorescence and thermographic images of hydrogel patches. (c) Thermographic surveillance of the photothermal heating of hydrogel patches in mice (nD5) 72 h after the first NIR treatment. Reprinted by permission from Springer Nature, ref. 99. Copyright 2016, Macmillan Publishers Limited. http://www.nature.com/nmat/ 177x108mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 82 of 84
Page 83 of 84 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 10. (a) Scanning electron microscope (SEM) and transmission electron microscopy (TEM) images of PLLA and Bi2Se3/PLLA membranes. (b) Infrared thermographic maps and time-dependent temperature increase in the tumor-bearing nude mice irradiated by an 808 nm laser. (c) Temperature growth during the irradiation of PLLA and Bi2Se3/PLLA membranes. (d) Corresponding growth curves of tumors in different groups of mice after NIR laser irradiation. Reprinted with permission from ref. 127. Copyright 2017 American Chemical Society. 177x149mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
TOC 43x28mm (600 x 600 DPI)
ACS Paragon Plus Environment
Page 84 of 84