You are on page 1of 27

Biomaterials 237 (2020) 119827

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Review

Advances in nanomaterials for photodynamic therapy applications: Status T


and challenges
Jianming Chena, Taojian Fana, Zhongjian Xiea, Qiqiao Zengb, Ping Xuec, Tingting Zhengd,
Yun Chend, Xiaoling Luob,∗, Han Zhanga,∗∗
a
Institute of Microscale Optoelectronics, Collaborative Innovation Centre for Optoelectronic Science & Technology, Key Laboratory of Optoelectronic Devices and Systems of
Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Micro-Nano Photonic Information
Technology, Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen University, Shenzhen, 518060, PR China
b
Department of Ophthalmology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen City, Guangdong Province, 518020, PR China
c
Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
d
Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China

A R T I C LE I N FO A B S T R A C T

Keywords: Photodynamic therapy (PDT), as a non-invasive therapeutic modality that is alternative to radiotherapy and
Photodynamic therapy chemotherapy, is extensively investigated for cancer treatments. Although conventional organic photosensitizers
Nanomaterial (PSs) are still widely used and have achieved great progresses in PDT, the disadvantages such as hydrophobicity,
Photosensitizer poor stability within PDT environment and low cell/tissue specificity largely limit their clinical applications.
Drug delivery
Consequently, nano-agents with promising physicochemical and optical properties have emerged as an attractive
2D nanomaterial
alternative to overcome these drawbacks of traditional PSs. Herein, the up-to-date advances in the fabrication
and fascinating applications of various nanomaterials in PDT have been summarized, including various types of
nanoparticles, carbon-based nanomaterials, and two-dimensional nanomaterials, etc. In addition, the current
challenges for the clinical use of PDT, and the corresponding strategies to address these issues, as well as future
perspectives on further improvement of PDT have also been discussed.

1. Introduction: photodynamic therapy for cancer treatment radiotherapy or chemotherapy in order to remove the cancer cells
completely [10], and it may suffer from recurrence for certain tumors.
In recent years, over 10 million new cancer cases are diagnosed Chemotherapy has an impact on cell division, thus associating with
every year, and around 9.6 million patients died from the disease in some side effects like myelosuppression, mucositis, alopecia, etc. [11].
2018 [1,2]. This number is projected to rise by about 70% over the Radiotherapy, on the other hand, needs to be operated precisely to the
following two decades [1,3]. In fact, cancer is one of the major causes of tumor site to minimize the damage to the surrounding normal tissues.
disease-related deaths, which accounts for a large percentage of total In the case of immunotherapy, the therapeutic outcome can be incon-
deaths (around 17% in 2018) [1]. Therefore, cancer treatment is a sistent for different patients [9,12]. In this sense, it becomes essential to
global task that shows a great impact to the whole society [4,5]. Cur- develop alternate treatment modalities that are less invasive, more ef-
rently, different types of clinical treatments for cancer are being fective, and more acceptable.
adopted, including surgery, radiotherapy, chemotherapy, and more Photodynamic therapy (PDT) is a potent candidate of such ther-
recently immunotherapy and molecular targeted therapy. In the past apeutic method which is rapidly developing for the diagnosis and
decade, the treatment programs with a combination of different treat- treatment of cancer. It is a non-invasive treatment strategy based on the
ment strategies, such as radiotherapy combined with immunotherapy, administration of a photosensitizer (PS), which can accumulate at the
chemotherapy combined with immunotherapy and molecular targeted tumor tissues and can be activated by a light source with a specific
therapy combined with immunotherapy, have also gained a lot of wavelength, resulting in the formation of cytotoxic reactive oxygen
progresses [6–9]. However, these conventional treatment modalities species (ROS) in the presence of endogenous molecular oxygen to
still show some drawbacks. For instance, surgery needs to work with eventually cause the death of cancer cells [4,13]. Importantly, the


Corresponding author.
∗∗
Corresponding author.
E-mail addresses: LXL2603@vip.sina.com (X. Luo), hzhang@szu.edu.cn (H. Zhang).

https://doi.org/10.1016/j.biomaterials.2020.119827
Received 25 October 2019; Received in revised form 13 January 2020; Accepted 25 January 2020
Available online 27 January 2020
0142-9612/ © 2020 Elsevier Ltd. All rights reserved.
J. Chen, et al. Biomaterials 237 (2020) 119827

individual component of PDT is non-toxic, and the generation of cyto- taking place depends on the specific PS used [22]. Hence, the PS with
toxic ROS, such as singlet oxygen (1O2), superoxide anion (O2•-) and high production ability of oxygenated products and singlet oxygen is of
hydroxyl radicals (%OH), only takes place when the PS is irradiated by pivotal importance for effective PDT [5].
light. In this regard, PDT can destroy the tumor cells in a safe manner, Many types of PSs have been developed for the treatment of cancer,
since the PS drugs themselves are minimally toxic without the presence which can be classified into different generations. Hemetoporphyrin
of external photo-activating light [14]. Moreover, PDT presents an ef- was found to be localized in tumor tissues in the early 1950's [23],
fective cancer treatment modality that can significantly reduce the side which motivated the purification and chemical modifications of he-
effects and improve the target specificity compared to the conventional matoporphyrin-based PSs for PDT. In the 1970's, Photofrin (structure
cancer treatment options like radiotherapy and chemotherapy, as only shown in Fig. 2) that was isolated from hemetoporphyrin mixture has
the target cells/tissues under light irradiation are treated [13,15]. A been approved by FDA for the use in cancer treatment [17]. The he-
variety of light sources like monochromatic light from laser sources matoporphyrin derivative (HpD) is considered as the first generation
(such as metal vapour lasers and argon-pumped dye lasers) or broad- PSs and was widely used in cancer therapy. However, the clinical use of
band light from non-laser sources (such as fluorescent and xenon arc the first generation PSs also suffers from several dilemmas: (1) they are
lamps) can be adopted for PDT [16]. The selection of the light source is generally excited by short wavelengths (such as visible light) that have
dependent on the PS used, tumor location and light dose. Obviously, poor penetration depth in tissue. Hence, their clinical application in
appropriate wavelengths that can excite the PS to produce ROS are PDT is limited for superficial tumors [24–27]; (2) synthesizing these PSs
required to induce PDT. The first discovery of PDT can date back to is rather complex and it is very difficult to purify them; (3) Photofrin is
more than 100 years ago, and the first modern application was reported not soluble in water, which also limits its biological use; (4) the molar
by Dougherty and co-workers in 1975 [17]. Since then, tremendous extinction coefficient is low, meaning high administration dosage of PS
investigations have emerged in this field and have proven PDT to be an is required for effective photodynamic activity. Additionally, they have
effective therapeutic modality for a variety of cancers, such as super- long half-life and tend to accumulate in the skin, causing skin photo-
ficial bladder cancer [18], head and neck cancers [19], and skin cancer sensitization for prolonged period [3,4].
[20]. The disadvantages of the first generation PSs triggered further re-
search on the second generation PSs with near IR (NIR) activation and
2. Principle of photodynamic therapy and different generations of high production of 1O2. The NIR light is known as the “window of
photosensitizers optical transparency”, which has minimal interaction with surrounding
biological components, hence affording increased tissue penetration
PDT is a combination of the photophysical and photochemical depth and high biomedical imaging resolution [28,29]. Typically, these
processes, which eventually leads to biological effect [5]. Fig. 1 illus- are macrocyclic compounds derived from substitutions of the porphyrin
trates the principle of PDT. Initially, the PS in ground state is trans- moieties or direct modifications of the porphyrin core, or some new
formed into an excited singlet state (1PS*) upon absorption of light. non-porphyrinoid PS molecules, including phthalocyanines [30–34],
This PS in its excited singlet state has very short lifetime, and may re- chlorins [35], bacteriochlorins, porphycenes [36,37], hypericin
turn to the ground state by means of releasing light energy (fluores- [38–40], phenothiazines (methylene blue and toluidine blue) [41–43],
cence) or heat energy (non-radiative decay). Alternatively, the multi- cyanines (merocyanine-540) [44,45], xanthenes (rose bengal) [46], and
plicities of the PS are possibly changed from singlet to triplet state metalated derivatives of PS (such as metalloporphyrins, AlPcS4, Si(IV)-
(3PS*) that has a relatively longer lifetime through intersystem naphthalocyanine, and tin ethyl etiopurpurin) [4,47–50], etc. (Fig. 3).
crossing. Similarly, the triplet state PS may also return back to the Nevertheless, the main drawback of the second generation PSs is their
ground state via the emission of phosphorescence or the release of heat non-specific localization at targeted cells/tissues, therefore the atten-
energy. More importantly, this triplet excited PS can react with cellular tion was turned to design the targeted third generation PS.
substrates directly via electron transfer, which leads to the formation of Generally, targeting agents are conjugated to the PS to enhance the
free radicals that can generate oxygenated products (e.g., O2%-, %OH and tumor targeting abilities (some examples shown in Fig. 4) [51–53].
H2O2) after reaction with O2 (Type I reaction). Alternatively, it can Alternatively, tumor specific carriers can be utilized to deliver the PSs
transfer energy to oxygen (3O2) to produce highly reactive 1O2 (Type II to the target site [4,5]. However, the tumor targeting efficiency is still
reaction). The reactive species formed during the photodynamic pro- not high enough for clinical uses [54], and organic PSs mostly suffer
cess can eventually result in killing cancer cells directly by means of from stability issues, resulting in reduced reaction rates [55].
apoptosis, necrosis or autophagy, and indirectly by damaging tumor Despite some of early generation PSs have been approved for clin-
vasculature that leads to tumor ischemia. Additionally, PDT is able to ical use (Table 1), the shortcomings, like hydrophobicity and low pe-
elicit immune response against tumor antigens [13,15,21]. PSs can use netration depth of the first generation PSs, and low cell/tissue specifi-
either type I or type II reaction, or both of them simultaneously to city associated with the second generation PSs, still hinder their clinical
destroy cancerous cells. The proportion of these two types of reactions therapeutics. Over the past few years, extensive attention has been paid
to the investigation of nanomaterial-based PDT, which presents a new
therapeutic modality that uses nanomaterial as a carrier or PS. Com-
pared to the organic PSs, nanomaterials represent some unique prop-
erties that make them more potent for PDT. First of all, nanomaterials
are stable under irradiation. Next, nanomaterials have very promising
optical properties which lead to improved penetration and efficacy of
PDT [22]. Moreover, molecule-modified nanocarriers are capable of
delivering PS to the target cells more precisely than traditional PSs,
transferring into improved curative effect and reduced side effects
[56,57]. Some nanomaterials, such as semiconducting nanoparticles,
are able to produce ROS intrinsically via an energy transfer or electron
transfer process, owing to their particular light absorption properties
[58]. Nanomaterials are more widely utilized as a carrier for PS, which
allows for targeting delivery of PS toward the tumor site with increased
drug loading efficiency and enhanced uptake by the cancer cells, due to
Fig. 1. Modified Jablonski diagram showing the principle of PDT. the large surface area and versatile surface modifications [4]. In this

2
J. Chen, et al. Biomaterials 237 (2020) 119827

NaO2C(H2C)2 Me R Me Me R

N Me Me N
N Me Me
NaO2C(H2C)2 Me O H
NH NH N N HN
N
Me H O OC(CH2)2 H Me Me (CH2)2CO2Na
N N N
Me

R Me NaO2C(H2C)2 Me Me (CH2)2CO2Na
n

Me
R= or n = 0-6
OH

Fig. 2. Structure of Photofrin®.

review, some of recent progresses on the use of nanomaterials in PDT, mostly exhibited PDT effect when excited by a 980 nm light, whereas
including various types of nanoparticles, carbon-based nanomaterials, 808 nm irradiation led to effective PTT effect. Upon 940 nm excitation,
and two-dimensional nanomaterials, etc., will be reviewed. Further- in vivo studies demonstrated that the gold nanoshells can trigger bi-
more, special interest is devoted to the current challenges of PDT and modal PDT/PTT for more efficient treatment of B16F0 melanoma tumor
the corresponding actions to deal with these issues. In the last section, than the clinically used drug doxorubicin. Of note is that, no photo-
future perspectives for the viable application of PDT will be discussed sensitizing drugs have been approved by FDA for melanoma cancer yet.
by exemplifying the multimodal treatment modality and several ad- By using gold bipyramids, He et al. found that singlet oxygen can be
vanced techniques. generated upon irradiation with a wide range of wavelengths
(660–975 nm) [65]. Particularly, the highest formation of 1O2 was
obtained when the gold bipyramids were excited at the wavelength
3. Application of nanomaterials in photodynamic therapy overlapping with the localized surface plasmon resonance (LSPR) peak.
This observation was very different from that of other gold nano-
3.1. Gold nanoparticles particles. In addition, it was demonstrated that efficient formation of
singlet oxygen can be achieved under low-intensity light irradiation
Gold nanoparticles have been investigated for many years to induce (200 mW/cm2).
PDT due to some of their promising properties, such as high surface Generally, current PDT uses one-photon absorption to excite PS to
areas, good biocompatibility and facile surface modification via gold- produce ROS. In earlier studies, metal nanoparticles have shown the
thiol chemistry [59]. Additionally, gold nanoparticles are also ex- abilities to generate singlet oxygen via the one-photon excitation me-
tensively studied in diagnostic applications, due to their tunable optical chanism [66,67]. However, one-photon excitation may cause potential
scattering and absorption [14,60,61]. The first use of gold nanorods (Au photodamage to the adjacent tissues around tumor site. Therefore, two-
NRs) alone (without employing an organic PS) for PDT was reported by photon excitation with precise manipulation of treatment dose is more
the Hwang group in 2014 [62]. Under the excitation of NIR light preferable in this sense. In two-photon PDT, femtosecond laser beam is
(915 nm, λ1), gold nanorods were found to be able to produce singlet utilized to obtain high fluxes of light. In the work of Jiang and co-
oxygen leading to the destruction of B16F0 melanoma tumors in mice workers, aggregates of gold nanospheres and gold nanorods have been
(Fig. 5). Next to the generation of 1O2, the excitation of gold nanorods assessed as PSs for two-photon PDT [68]. Upon irradiation with fem-
with 780 nm (λ2) can also increase the temperature around tumor tosecond laser pulses at 800 nm, two-photon induced generation of
tissues by converting the photon energy to heat, as confirmed by the singlet oxygen was observed using either unaggregated or aggregated
formation of heat shock protein (HSP 70). In this process, gold nanorods gold nanoparticles. However, the 1O2 generation capability was gen-
mostly exerted the so-called photothermal therapy (PTT) effect, leading erally enhanced by aggregated gold nanoparticles. That is, the 1O2
to the apoptosis of cancer cells (Fig. 5b). By changing the activation generation capability by aggregated gold nanospheres was 8.3-fold
wavelengths (λ1 or λ2), the dominating phototherapeutic effects can be higher than that by the unaggregated ones. A similar trend was ob-
switched between PDT and PTT. In particular, further experiments served when aggregated gold nanorods were used, in which the effi-
showed that gold nanorods usually provided much higher PDT effect (at ciency of singlet oxygen generation was improved for 1.8 times with
least 10 folds) than the PTT effect in killing tumor cells. Additionally, it respect to the case of unaggregated gold nanorods.
was able to track the distribution of gold nanorods in vivo through the Along with the fast development of nanotechnology, there are
single photon‐induced fluorescence emitted by themselves. various synthetic methods available for the researcher to obtain the
In the same year, the same group reported the production of singlet gold nanoparticles with satisfactory architecture and features for PDT
oxygen by gold nanoechinus (Au NEs), a different morphology of gold application [69]. Next to the multiple physicochemical properties, the
nanoparticles, under excitation with wavelengths covering both the possibility of further chemical modifications enables the improvements
biological windows NIR I (650–950 nm) and NIR II (1000–1350 nm) in bioavailability and usability, making the gold nanoparticles a potent
(Fig. 6) [63]. The extinction coefficients of gold nanoechinus observed candidate for clinical cancer treatment.
at 915 nm (NIR I) and 1064 nm (NIR II) are both greater than that
previously observed from conventional organic PSs and gold nano-
particles. Owing to the extraordinarily high extinction coefficients of 3.2. Silver nanoparticles
these gold nanoechinus, it was able to significantly decrease the amount
of nanoparticles used, the light intensities, as well as the irradiation Silver is known as one of the strongest natural antibiotics and has
time for deep tissue cancer treatments. been used by humans to kill a variety of microorganisms for a long time.
The Hwang group also demonstrated that gold nanoshells, including Nowadays, silver nanoparticles are also widely used in antibacterial
nanocages, nanorod-in-shell and nanoparticle-in-shell, can produce treatment. One of the intriguing properties of the silver nanoparticles is
singlet oxygen and induce PDT as well for the complete removal of solid that they typically have an exceptionally high specific surface area,
tumors in mice [64]. Similar to the phototherapeutic effects induced by which means the contact surface area of silver with bacteria can be very
gold nanorods, it was possible to control and switch the dominant role large. Consequently, using silver nanoparticles would significantly re-
of PDT and PTT by changing the activation wavelength. Gold nanocages duce the amount of silver (up to hundreds of times) without a drop in

3
J. Chen, et al. Biomaterials 237 (2020) 119827

N
NH N
NH N NH N NH N
N N
N HN N HN N HN N HN
N

Phthalocyanine Chlorin Bacteriochlorin Porphycene

OH O OH

Cl
Me OH Cl Me2 N S NH2
Me2 N S NMe2
Me OH
N Me
N

OH O OH
Hypericin Methylene blue Toluidine blue

SO3Na Cl
Cl Cl
N N N
O Bu KO2C Cl H
M H
O N I I N N
S
N
KO O O
O Bu M = metal
I I
Merocyanine-540 Rose bengal Metalloporphyrin

HO3S SO3H CO2Et


Me Me
N N Me
Me
N N N N H
N Cl N
N Al Cl N N Si N Sn
N N N N
N Cl N
N N Me
Me
HO3S SO3H Me
Me
Aluminum phthalocyanine tetrasulfonate Tin ethyl etiopurpurin
(AlPcS4) Si(IV)-naphthalocyanine (SnET2)
Fig. 3. Structures of some second generation PSs.

bactericidal effects [70]. Silver nanoparticles are also found to show the band of the silver and gold nanoparticles, in order to sensitize the
ability to generate the singlet oxygen, thus attracting many efforts on formation of singlet oxygen. For example, photo-excitation of silver
their use in PDT. Same as the cases of gold nanoparticles, silver nano- decahedrons (ca. 520 nm LSPR band) at the NIR wavelength of 885 nm
particle-mediated sensitization and generation of singlet oxygen are resulted in a significant formation of singlet oxygen, in sharp contrast,
highly related to their morphology, as reported by Hwang et al. [71]. activating gold decahedrons that have a LSPR band in this wavenumber
For instance, while either a very small or no 1O2 phosphorescence range with the same laser light did not give any 1O2 phosphorescence
signal was observed when silver nanocubes or gold decahedrons were signal. Similarly, silver triangular plates (ca. 590 nm LSPR band) gave a
utilized, excitation of silver decahedrons and silver triangular nano- strong 1O2 phosphorescence signal under 544 nm excitation, whereas
plates can lead to the production of singlet oxygen. In general, it was irradiating silver nanocubes (ca. 500 nm LSPR band) at 525 nm only
found that the radiation wavelength should be different from the LSPR resulted in weak generation of singlet oxygen.

4
J. Chen, et al. Biomaterials 237 (2020) 119827

N OR N O O
O Si R=
H N N O
N OMe O O
HNAc N OR N
O O N

Ac-L-Phe-ALA-OMe
Galactose substituted Si(IV)-phthalocyanine

R
F F OH
HO
F O S
F
HO OH
F F F F
NH N
or
R R R=
N HN OH
F F F F
O S
F F HO
HO OH

F F
R
R
F F
OH OH

F F N HO
OH OH
O OH
F F F F
NH N S HO
R R or OH O O OH
HN R=
N
F F F F HO
OH O O OH
F F
HO
O OH
F F
R S
Suger conjugated chlorins
Fig. 4. Some examples of third generation PSs (conjugated with targeting agents) [51–53].

The diverse morphologies and compositions of silver nanoparticles respectively. Organically modified silica (ORMOSIL) nanoparticles can
can translate into highly tunable optical features, endowing them with be conjugated with both hydrophilic and hydrophobic molecules effi-
attractive applications in PDT [72]. Moreover, silver nanoparticles have ciently, which makes them promising to transport PS drugs [75]. In
the ability to cause oxidative stress to tumors, leading to the apoptosis 2007, Ohulchanskyy and co-authors incorporated PS molecules into
of tumor cells [73]. ORMOSIL nanoparticles covalently, finding that the covalently bound
PS molecules could generate singlet oxygen robustly upon excitation
while retaining their spectroscopic and functional properties [76]. In
3.3. Silica and silicon nanoparticles
this study, an ORMOSIL precursor was incorporated with a PS iodo-
benzylpyropheophorbide (IP), forming iodobenzyl-pyro-silane (IPS).
Although silica is not active for PDT itself, silica nanoparticles can
Subsequently, IPS was coprecipitated with the ORMOSIL precursor vi-
be used for encapsulating PSs in PDT, due to the nontoxicity, chemical
nyltriethoxysilane (VTES) to provide the ORMOSIL nanoparticles.
inertness and optical transparency of silica [22]. Additionally, it is
These as-prepared nanoparticles have a size of ca. 20 nm and show high
feasible to make chemical functionalization for silica, owing to the
monodispersity and good stability in aqueous solution. It was demon-
hydroxyl groups on the silica surface. In this regard, silica nanoparticles
strated in vitro that the ORMOSIL nanoparticles can be uptaken by
are normally used for drug delivery in the studies [22]. In 2007, Yan
tumor cells effectively and were capable of killing colon carcinoma cells
and co-authors embedded the PS meta‐tetra(hydroxyphenyl)‐chlorin
upon irradiation at 514 nm.
(m‐THPC) into sol–gel silica matrix-based nanoparticles via a modified
Mesoporous silica nanoparticles (MSNs) have also been extensively
Stöber sol–gel process [74]. It was shown that this Si/m‐THPC nano-
utilized for delivering PS due to the intriguing features like large sur-
system produced singlet oxygen more efficiently than the free THPC
face area and pore volume, and high chemical stability [77,78]. MSNs
molecules, i.e., the exact reaction rate constants with anthracene‐9,
can be used as a nanocarrier for hydrophobic PSs, such as ZnPc that
10‐dipropionic acid for free m‐THPC and m‐THPC embedded in silica
tends to self-aggregation in aqueous medium. In 2012, Tu et al.
nanoparticles were 2.7 × 108 M−1 s−1 and 4.8 × 108 M−1 s−1,

5
J. Chen, et al. Biomaterials 237 (2020) 119827

Table 1
Some first and second generation PSs approved for clinical use.
PS Tradename Chemical structure Indication uses Maximum Extinction
absorption coefficient (M−1
wavelength cm−1)

HpD Photofrin See Fig. 2 Lung, esophageal, bladder, 630 3000


gastric cancer
5-Aminolevulinic acid (5-ALA) Levulan O Actinic keratoses, basal cell 635 < 1000
carcinoma, head and neck
H 2N OH
cancer
O
5-ALA-hexylester Hexvix O Diagnosis of bladder cancer 400 < 1000
H 2N O

O
Methyl aminolevulinate (MLA) Metvix O Actinic keratoses, basal cell 635 < 1000
H 2N O carcinoma

O
Meta-tetrahydroxy phenyl Foscan OH Prostate and pancreatic tumors, 652 30,000
chlorine (m-THPC) head and neck cancer

OH
N
H
N N
H
N
HO

HO
Sulfonated Aluminium Photosens See Age-related macular 675 110,000
phthalocyanine (AlPcS4) Fig. 3 degeneration, prostate cancer
Benzoporphyrin derivative mono- Visudyne Age related macular 693 35,000
acid (BPD-MA) degeneration, non-melanoma
skin cancer

Mono-L-aspartyl chlorine e6 Aptocine/ Lung cancer, recurrent 664 45,000


(NPe6) Laserphyrin subcutaneous tumors

incorporated polyethyleneglycol (PEG)- and polyethylenimine (PEI)- Importantly, injection of PEG-PEI-MSNs/ZnPc resulted in efficient
modified zinc phthalocyanine (ZnPc) to MSNs, forming a PEG-PEI- passive tumor targeting and successful destruction of tumor upon ex-
MSNs/ZnPc nanosystem that was developed for PDT [79]. In this study, posure to 680 nm light (Fig. 7b–c). MSNs functionalized with galactose
PEI was used to facilitate the endosomal escape of MSNs from the ly- on the surface were reported in the same year for the use as delivery
sosome to the cytosol via a “proton sponge” mechanism. However, the vehicle and PDT agent [80]. Confocal microscopy experiments revealed
incorporation of PEI also introduced the side effect of high cytotoxicity that galactose functionalization can contribute to the enhanced uptake
aroused by its cationic property. Therefore, PEG was used to further by the cancer cells through galactose receptor–mediated endocytosis,
modify PEI-MSNs to reduce the toxicity and enhance the biocompat- resulting in the accumulation of the nanoparticles in the endosomal and
ibility. By doing so, the phototoxicity of the PEG and PEI modified lysosomal compartments. The MSNs equipped with a PS (porphyrin)
MSNs/ZnPc was largely improved in vitro, in comparison with the solely and an anti-cancer drug (camptothecin) were tested for synergistic
ZnPc-loaded MSNs. As shown in Fig. 7a, all of the nanoparticles showed anticancer effects, which showed a dramatically increased death of
highest biodistribution at tumor site, and the PEGylated MSNs/ZnPc cancer cell under light at 650 nm compared to separate treatments.
exhibited enhanced uptake in tumor over unmodified ones. In 2017, folic acid (FA)-modified silica nanoparticles were prepared

6
J. Chen, et al. Biomaterials 237 (2020) 119827

(PDA) layer and PEG-FA decoration [82]. Under 808 nm excitation, the
PDA layer and ICG were able to exert PTT effect, and ICG was capable
of producing ROS as well. Additionally, the light irradiation can also
mediate the vaporization of PFH to generate bubbles for tumor ultra-
sonic (US) imaging and ICG can provide NIR fluorescence emission
upon release. Consequently, combined PTT/PDT mediated effective
inhabitation of MCF-7 tumors in mice guided by UC/fluorescence
imaging has been achieved in vivo by this MSNs nanosystem. Very re-
cently, a tumor-targeting PDT based on zinc(II) phthalocyanine deri-
vative (PcC4)-incorporated MSNs and a wrapping DNA has been re-
ported by Li et al. [83]. The selectivity was realized by the self-
quenching photoactivity of PcC4 and the subsequent recover of pho-
toactivity driven by the reaction between the nanocomposites and
proteins (telomerase and albumin). In the study of Lin et al., ZnPc was
conjugated to MSNs via an acid-responsive hydrazone bond [84]. The
as-obtained ZnPc-MSNs nanoplatform showed no photodynamic ac-
tivity due to the aggregation of PS ZnPc. Nevertheless, the generation of
singlet oxygen can be selectively turned on through the release of PS
upon acidic solution treatment.
Porous silicon nanoparticles were found to be able to generate
singlet oxygen under light covering entire visible range, due to broad
absorption band of porous silicon [85]. Xiao et al. etched single crystal
silicon wafers electrochemically to prepare porous silicon nano-
particles, which can produce singlet oxygen with a quantum yield of
~0.1 or ~0.17 in ethanol or H2O2, respectively. Under white light il-
lumination, these Si nanoparticles were proven to cause significant cell
death in vitro [86]. More recently, porous silicon nanovectors with gold
Fig. 5. Schematic working mechanisms of gold nanorod‐induced PDT/PTT ef- nanoparticle decoration and mannose tumor targeting agent functio-
fects under NIR laser light. (a) Destruction of melanoma tumors in mice in vivo nalization were reported by Chaix et al. [87]. The as-prepared silicon
by Au NRs. (b) Various cellular events involved in cellular deaths induced by nanostructures showed phototoxicity toward human breast cancer cells
gold nanorod‐mediated PDT/PTT effects upon photoexcitation. Reproduced under NIR light excitation. Porous silicon nanorods have also been
with permission [62]. Copyright 2014, Wiley-VCH Verlag GmbH & Co. KGaA. demonstrated to be capable of inducing photodynamic effects with a
quantum yield of singlet oxygen at 0.24 [88]. Accordingly, decreased
by Bharathiraja et al. for site-specific delivery of PS chlorin e6 (Ce6) viabilities of HeLa cells cultured with these silicon nanorods were ob-
[81]. Efficient generation of singlet oxygen was obtained with 670 nm served from in vitro assays with 635 nm laser exposure.
irradiation, leading to improved killing efficacy of MDA-MB-231 cells Using different synthetic methods, a variety of silica and silicon
than free Ce6. Very recently, a multifunctional nanoplatform based on nanoparticles with preferable size and porosity can be prepared for
perfluorohexane (PFH)-encapsulated MSNs was fabricated by Huang PDT. Besides, it is possible to incorporate cancer-specific agents to the
et al., with PS indocyanine green (ICG) loaded within polydopamine silicon-based nanoparticles via surface functionalization, hence

Fig. 6. Au NEs-induced PDT effects illustrated by the


changes of tumor volume after treatment with dif-
ferent conditions for 14 days, and photos of tumor-
bearing mice at the 14th day in different groups
under irradiation with NIR I (808 and 915 nm) and
NIR II (1064 nm) Reproduced with permission [63].
Copyright 2014, Wiley-VCH Verlag GmbH & Co.
KGaA.

7
J. Chen, et al. Biomaterials 237 (2020) 119827

amount of porphyrin, which was attributed to the increased effective


overlap integral due to the interaction between multiple porphyrin
acceptors and a central QD donor [95]. A water-soluble QD-organic dye
nanosystem has been used by Shi et al. as a PS for PDT, which was
synthesized from CdTe QDs, the stabilizer 2-aminoethanethiol and the
organic dye meso-tetra(4-sulfonatophenyl)porphine dihydrochloride
(TSPP) [96]. The surface of the CdTe QDs was modified with TSPP via
electrostatic interaction, as a result, slight shift in the luminescence
maximum to the blue region was observed, as compared to the case
with free TSPP. Production of singlet oxygen was obtained when irra-
diating the CdTe QD-TSPP with near-UV or visible light. A significant
amount of singlet oxygen (0.43 quantum yield) was detected using a
355 nm Nd:YAG laser where the absorption of TSPP is minimal
(~10%). In addition, no phosphorescence signal from singlet oxygen
was detected when the CdTe QDs were irradiated without the presence
of TSPP. Therefore, it indicated that singlet oxygen was produced from
the irradiation of the QDs followed by a FRET-type mechanism. In
2015, water-soluble nanocomposites containing ZnSe/ZnS QDs and Ce6
were synthesized by Martynenko et at. For PDT in cancer cells [97].
Approximately 50% intracomplex FRET between ZnSe/ZnS QDs and
Ce6 was detected, indicating these nanocomposites were an effective
photosensitizing agent. PDT test was done by employing the nano-
Fig. 7. (a) Biodistribution of different MSNs/ZnPc nanoparticles in H22 tumor composites against the Erlich acsite carcinoma cells, and it was revealed
bearing mice. (b) Changes of tumor size in different groups as a function of that the cancer cell photodynamic destruction was enhanced for two
time. (c) Images of mice bearing H22 tumor after treatment with different times in comparison with that of free PS Ce6. This significant en-
conditions for different experiment time. Reproduced with permission [79]. hancement was attributed to the efficient photoexcitation energy
Copyright 2012, Elsevier.
transfer from QDs to Ce6, and the enhanced cellular uptake of the PS
Ce6 incorporated to ZnSe/ZnS QDs.
improving the tumor-targeting ability [78]. Moreover, recent studies More recently, Shen and co-authors reported a QD-based tumor-
have revealed their promising uses in NIR and two-photon PDT for deep targeting hybrid nanocomposite, in which a QD-Zn-porphyrin (TMPyP-
cancer therapy. Zn-QD) nanocomplex, a highly fluorescent PS rhodamine 6G (R6G) and
a NIR fluorophore NIR775 were encapsulated in FA decorated phos-
3.4. Quantum dots (QDs) pholipid polymers (Fig. 8) [98]. Porphyrin can be largely loaded on
these nanoparticles, and the light absorption capability was found to be
strong. As a consequence, a very high singlet oxygen quantum yield of
Another type of nanomaterials, QDs, have also been extensively
studied for the application in PDT. QDs possess some unique optical ca. 0.91 was achieved through an effective dual energy transfer process.
Additionally, the folate receptor can contribute to the highly selective
properties, including high emission quantum yield, size- and composi-
tion-tunable emission, as well as facile surface modification, that are delivery of as-prepared nanoparticles to cancerous tissues, thus al-
lowing non-invasive fluorescence imaging and successful photodynamic
important for being promising nanocarriers [89–91]. Moreover, QDs
are also utilized as donors in fluorescence resonance energy transfer destruction of tumors in vivo without scathing the surrounding healthy
cells. Zhang and co-workers revealed that Ag2S QDs can be utilized as a
(FRET), which is used extensively for studying the dynamics of biolo-
gical macromolecules [91–93]. potent fluorescent probe that have bright photoluminescence, good
biocompatibility, and emission in the NIR biological window II [99].
In 2010, biocompatible CdSe QDs conjugated with water-soluble
porphyrin were prepared by Qi et al. for PDT through two-photon ex- These Ag2S QDs showed strong targeting abilities and imaging of dif-
ferent cell lines in vitro, such as human breast cancer cell line and
citation [94]. It was concluded that porphyrin-conjugated QDs under
two-photon excitation provided 2-fold higher 1O2 quantum yield than human glioblastoma cell line. Additionally, it was illustrated from cy-
totoxicity study that the Ag2S quantum dots exhibited negligible toxi-
the solely porphyrin solution. By employing the FRET mechanism, en-
ergy was transferred efficiently between the QDs and the porphyrin. city in the aspects of cell proliferation, apoptosis and necrosis, ROS
generation, as well as DNA damage. It is important to use external
Moreover, the energy transfer efficiency was enhanced by the larger

Fig. 8. Illustration of the fabrication of the hybrid nanocomposite contain the TMPyP-Zn-QD nanocomplex, the PS R6G, NIR fluorophore NIR775 and phospholipids,
and their use in tumor selective imaging and PDT. Reproduced with permission [98]. Copyright 2017, Elsevier.

8
J. Chen, et al. Biomaterials 237 (2020) 119827

Fig. 9. (a–b) Injection of FA-PEG-UCNPs into the mice bearing melanoma tumors and time-dependent changes of tumor volume under different conditions. The
group with the treatment of FA-PEG-UCNPs showed greatest tumor growth suppression. (c) Images of the mice under different conditions (FA-PEG-UCNPs, UCNPs
alone or PBS) showing the changes of tumor volume before and after PDT treatment (at the 7th day). Reproduced with permission [110]. Copyright 2012, Nature
Publishing Group.

excitation light source with proper tissue-penetrating properties to ac- activation wavelengths of these PSs are within the range (< 700 nm)
tivate PS drugs, in order to achieve successful clinical application of where endogenous molecules (like hemoglobin) have strong absorp-
PDT. In 2012, Hsu and co-authors developed a new type of QD con- tion, which largely limited their use in PDT. In this study, the genera-
jugate immobilized with light-emitting Renilla luciferase 8 (QD-RLuc8), tion of singlet oxygen mediated by the MC-540-coated UCNPs was
which can produce bioluminescence as the external light source [100]. successfully detected with NIR excitation by measuring the decrease in
The addition of coelenterazine triggered the bioluminescence resonance the fluorescence bands of the 1O2 sensor 9,10-anthracenedipropionic
energy transfer (BRET) from the substrate to the QD conjugates, acid (ADPA). In vitro PDT tests showed that the as-obtained MC-540-
thereby inducing self-illumination at 655 nm to activate the PS m-THPC coated UCNPs exhibited photodynamic cytotoxicity to MCF-7/AZ
(Foscan®) to exert PDT effects. It was shown that QD-RLuc8 with coe- breast cancer cells. UCNP-mediated PDT can also be used for in vivo
lenterazine could sensitize efficient production of ROS by using BRET- tumor therapy, as first demonstrated by Idris and co-authors [110], who
mediated PDT, resulting in killing half amount of A549 cells with used NaYF4:Yb/Er nanoparticles coated with mesoporous silica as a
Foscan® in vitro and remarkably suppressed tumor growth in vivo. These nanotransducer and a carrier of two different PSs (i.e., ZnPc and MC-
results demonstrated that it is able to realize deep tumor therapy by 540). Improved ROS production ability and reduced cell viability were
using BRET-mediated PDT. obtained, showing that the synthesized UCNPs with the dual-PS gave
To date, a wide range of different QDs have been exploited for PDT higher PDT efficacy compared to that using a single PS. Both direct
owing to their promising characteristics, providing many achievements injection of the UCNPs into melanoma tumors and intravenous injection
in cancer treatment. Nevertheless, the major drawback of using QDs is of FA and PEG modified UCNPs (FA-PEG-UCNPs) into tumor-bearing
their potential toxic effects, which is mainly due to the absorbed or- mice have led to tumor growth inhibition in vivo upon 980 nm excita-
ganic species on the surface from the synthesis stage [101], and the tion. In the latter case, the tumor targeting capabilities and the circu-
release of heavy metals from the QD cores (such as CdSe and CdS) latory lifetime of the UCNPs were improved by FA and PEG, respec-
caused by surface oxidation upon oxygen/UV exposure [102–104]. In tively, which resulted in more significant PDT effects, as shown in
this regard, continuous efforts should be paid on developing QDs with Fig. 9.
less toxicity, in order to achieved clinical PDT applications. NaYF4:Yb/Er nanoparticles coated with polymer have also been
used as a delivery vehicle for the PS Ce6, which formed a UCNP–Ce6
3.5. Upconversion nanoparticles supermolecule complex [111]. This UCNP–Ce6 nanosystem showed two
emission bands at 550 and 660 nm upon 980 nm irradiation. Of note is
The upconversion nanoparticles (UCNPs) are able to convert low that the 660 nm emission wavelength overlaps with the absorption
energy NIR light into higher energy visible/UV light using a non-linear band of Ce6. Singlet oxygen production and good PDT performance
anti-stokes mechanism [105,106]. During PDT within the biological were achieved with 4T1 murine breast cancer cells upon intratumoral
environment, this emitted photon can activate the PS to sensitize the injection of UCNP–Ce6 under NIR light irradiation. Notably, there was
formation of ROS. Generally, the photon upconversion process involves nearly no observation of the administrated UCNPs in the mouse body
a real metastable electronic state of lanthanide ions embedded into a after 1–2 months, which demonstrated their nontoxicity to the treated
host lattice with a lifetime scale of microsecond. Whereas two-photon animals. Chen et al. doped gadolinium (Gd)‐ion to the NaYF4:Yb/Er
PDT requires simultaneous absorption of two NIR photons involving a UCNPs to fabricate the NaYF4:Er/Yb/Gd upconversion nanocrystals
virtual state with an ultra-short lifetime (femtoseconds). Therefore, the [112]. By using a water‐in‐oil reverse microemulsion strategy, the hy-
conversion efficiency of upconversion process is orders of magnitude drophilic PS drug methylene blue (MB) was efficiently conjugated to
higher than that of a two-photon excitation mechanism. Hence, a much the UCNPs in a silica matrix, providing the UCNPs/MB nanocomplex
lower power density is required for achieving photon upconversion having a particle size smaller than 50 nm. The as-obtained UCNPs/
(typically 0.1–100 W/cm2) [107]. Additionally, the emission band of MB‐based PDT drug successfully generated singlet oxygen under
the UCNPs is similar to the absorption band of PS, resulting in improved 980 nm excitation. In contrast, no signal was observed in the case of
ROS production efficiency [22]. In this regard, the UCNPs can also act either using NaYF4:Er/Yb/Gd alone, or free MB solutions under the
as a promising carrier for anti-cancer drugs or PS in PDT. same conditions.
In 2007, Zhang and co-authors reported the use of the UCNPs in The reason for typically using NaYF4 crystal as host co-doped with
PDT for the first time [108]. The UCNPs used in this study, Yb3+/Er3+ in UCNP-based PDT, as discussed above, is that the emis-
NaYF4:Yb3+/Er3+, showed strong emission bands in the visible region sion bands of both Er3+ overlaps with the activation wavelengths of
around 537 and 635 nm when excited by an IR source (974 nm). The PS several organic and inorganic PSs [113]. Similar phenomenon was also
molecule, Merocyanine 540 (MC-540) was attached to the UCNPs observed when NaYF4 was doped with Yb3+/Tm3+. In 2017,
during the silica coating process. Together with rose bengal, Mc-540 Nd3+‐sensitized NaYF4:Yb/Tm was synthesized by Chan et al. for in vivo
has been used for generating ROS in the 1980's [109]. However, the PDT [114]. In combination with an inorganic PS, graphitic carbon

9
J. Chen, et al. Biomaterials 237 (2020) 119827

nitride quantum dots (CNQDs), this nano-pharmaceutical platform al- forming the core–satellite structures. In this system, the gold nanorod
lowed the absorption of 808 nm NIR light to produce UV light emission, dimer core can generate heat upon irradiation at 808 nm, thus acting as
which in turn produced ROS to achieve PDT effect. Consequently, high photothermal agent, whereas the UCNPs excited Ce6 to trigger PDT
energy light generated by UCNPs without overheating the tissue can be under excitation with 980 nm light. Furthermore, successful tumor
achieved due to the 808 nm excitation bands. Moreover, the conjuga- elimination was achieved without regrowth after combined photo-
tion of polymer poly(L‐lysine) (PLL) can improve the binding between therapeutic treatment with a low power dosage (2 mW/cm2 for 980 nm
the UCNPs and the CNQDs, as the surface of the nanoparticles was and 0.2 W/cm2 for 808 nm, respectively).
converted to full positive. Dou et al. coated the NaYF4: Yb/Tm UCNPs Next to the outstanding optical characteristics and rich surface
with a nanometric layer of silica, which were further modified with (3- functionalities, the energy transfer ability of UCNPs offers the oppor-
aminopropyl) triethoxysilane (APTES) using Stöber method [115]. tunity to achieve NIR-triggered deep tissue PDT while using appropriate
Subsequently, the UCNPs were covalently incorporated to the PS Ce6 energy to sensitize efficient formation of ROS. Hence, UCNPs present as
through the amino groups on the silica layer. The PDT effect was as- a powerful nanoplatform for PDT. Yet, the generally low upconversion
sessed with the MCF-7 human breast adenocarcinoma cell line in vitro. quantum yield (< 3%) and the relatively poor compatibility in phy-
By using a low concentration (50 μg/mL) of this UCNP–Ce6 nano- siological environment still hinder their biological application
composite, 50% of the cells were killed under mild dosage (7 mW/cm2) [107,124]. Besides, the potential side effects of emitted UV light, like
of 980 nm light for 10 min. In addition, more than 90% of cell viability photocarcinogenesis, should be carefully assessed before they can be
was achieved under the same conditions, indicating that the toxicity of applied in clinical use.
this UCNP–Ce6 nanosystem was low in the effective concentration
range. Alternatively, the UCNPs LiYF4: Tm3+/Yb3+ have also been used 3.6. Carbon-based nanomaterials
for singlet oxygen generation, which exhibited stronger blue emission
upon NIR irradiation, as compared to the NaYF4 UCNPs [116]. By using Carbon-based nanomaterials have aroused considerable research
m-THPC as PS, which was modified with 4-(bromomethyl)benzoic acid interests in the field of PDT for their inimitable optical and mechanical
(BMBA), the LiYF4: Tm3+/Yb3+- UCNPs produced singlet oxygen when features, versatile chemical functionalization, good biocompatibility
activated at 980 nm. In vitro studies demonstrated that this nanocon- and low toxicity [125–128]. Numerous carbon-based nanomaterials are
struct was able to kill HeLa cell from 20% to 70% under 980 nm irra- existing due to many allotropic forms of carbon. Amongst them, carbon
diation for 1 h. nanotubes (CNTs), fullerenes and graphene-based nanomaterials are
More recently, research attentions have been turned to focus on most widely applied in PDT.
investigating multifunctional UCNP-based nanocomposites that are
combined with image-guided PDT and multimodal therapy [117–119]. 3.6.1. Carbon nanotubes
Very recently, the UCNPs coated with the PS TiO2 have been adopted Carbon nanotubes (CNTs) can be considered as the one-dimensional
for image-guided in vivo PDT, which realized a complete photo- form of rolled-up single or multilayered graphene, with a typical inner
switching in UV–blue region [120]. The emission band of these UCNPs diameter between 1 and 10 diameters. Depending on the number of
depended on the wavelength of the excitation light, i.e., the prominent wrapped sheets, two types of CNTs, namely, single-walled carbon na-
UV–blue emission from Tm3+ was turned on or completely turned off notubes (SWCNTs) and multiwalled carbon nanotubes (MWCNTs), are
with 980 nm or 800 nm excitation light, respectively. Consequently, in usually studied. In addition to their exceptional physical properties, the
vivo tumor imaging prior to the actual treatment can be achieved. versatile surface modifications of CNTs can turn them into outstanding
Under 980 nm excitation, the Tm3+ emission band at 350 nm well PDT agents. Murakami et al. revealed that semiconducting and metallic
matched the absorption of TiO2, thus generating ROS to induce PDT. On SWCNTs were capable of producing ROS (singlet oxygen and O2%-)
the other hand, only the Er3+ emission band at 660 nm appeared under under NIR light irradiation (808 nm) [129]. It was found that SWCNTs
excitation at 800 nm, which allowed tracing the therapeutic process by exhibited stronger photodynamic effects than MWCNTs in this study.
monitoring the emission around 650 nm in vivo. More importantly, the death of cancer cells was achieved using
In recent years, the UCNPs have also been utilized in combination SWCNTs after stabilization with high-density lipoprotein. However, no
with other nanoparticles to realize multimodal tumor therapy, which in vivo experimental results on the destruction of solid tumors in animal
combines two or more therapeutic strategies of cancer (e.g. PTT, PDT, model have been reported in this study. Through functionalization of
radiotherapy, chemotherapy) [121]. It has been shown that different SWNTs with PEI covalently or polyvinylpyrrolidone (PVPk30) non-
therapeutic modalities can take effect synergistically, which leads to covalently, two modified SWCNTs were prepared by Wang and co-
improved anticancer outcomes, thereby attracting the interest in de- workers [130]. It was concluded that the functionalization method had
veloping nanomaterials that are able to induce multimodal therapy an impact on the photodynamic ability, and the PEI functionalization
[117,122]. In 2015, gold nanorod dimer-UCNP-Ce6 assembly was fab- showed higher photodynamic efficiency than the PVPk30 modification
ricated by Sun et al. to achieve combine PTT/PDT, which allowed both in vitro and in vivo. The SWCNTs have also been used for delivering
successful tumor therapy in vivo (Fig. 10) [123]. The gold nanorod PS drugs to induce photodynamic activity. In the work of Staicu et al.,
dimer (core) and the UCNPs NaGdF4 (satellite) loaded with the PS Ce6 PS verteporfin was loaded on the SWCNTs functionalized with amino
were hierarchically assembled via complementary base pairing, covalently for PDT [131]. In order to obtain higher quantum yield of
1
O2, verteporfin needed to be conjugated to the amino-SWCNTs rather
than mixed physically (51% vs. 23%). This was a result of the smaller
quenching of 1O2 due to the conjunction between verteporfin and the
amino-SWCNTs. Very recently, a multifunctional nanoplatform was
reported by Zhang et al., in which Fe3O4@CQDs were coated on PE-
Gylated SWCNTs [132]. By loading the chemotherapy drug doxorubicin
(DOX) and conjugating with sgc8c aptamer, this nanoplatform can be
used simultaneously for combined photodynamic/photothermal
therapy, drug release, as well as magnetic resonance imaging (MRI).

Fig. 10. Illustration of the fabrication of gold nanorod dimer-UCNP-Ce6 as- 3.6.2. Fullerenes
sembly for imaging-guided combinational phototherapy. Reproduced with Fullerenes are three-dimensional spherical, ellipsoidal or tubular
permission [123]. Copyright 2015, Wiley-VCH Verlag GmbH & Co. KGaA. structure of all-carbon materials, with C60 as the mostly studied

10
J. Chen, et al. Biomaterials 237 (2020) 119827

example. Although fullerenes alone are poorly soluble in water and they GO nanomaterials possess abundant oxygen-containing moieties on the
exhibit intrinsic toxicity, a variety of modifications on the surface of surface. These characteristics allow further modifications by many
fullerenes have been developed to overcome these issues [133]. In functional molecules, such as hydrophilic macromolecules, targeting
addition, fullerenes have been proven to possess higher biocompat- agents and active agents, thus extending their biological applications
ibility than other carbon-based materials like graphene and CNTs and reducing the toxicity [144,151]. Notably, the fluorescence
[134–137]. In 2015, Li et al. reported a fullerene-based multifunctional quenching ability of GO nanomaterials is extremely high, which further
nanocomposite, in which C60 was modified with phenylalanine and was expands the applications in PDT [152]. In 2016, Ding and co-workers
attached with polylactic acid (PLA) [138]. This as-prepared nanoplat- loaded the PS hypocrellin A (HA), and TiO2 nanoparticles onto the
form can highly selectively transport anticancer drug mitoxantrone to surface of GO, forming a light-sensitive drug delivery system [153].
the tumors in C57BL mice, therefore mediating combined che- Increased ability of generating ROS was realized through mutual sen-
motherapy/PDT effects upon visible light irradiation with negligible sitization mechanism, in which the sensitization effect was formed from
toxicity to the normal healthy organs. Later on, the same group re- the HA–TiO2 stable complex. Simultaneously, modifying TiO2 with GO
ported a drug delivery system adopting PEGylated C60 with a tunable could also generate ROS upon exposure to visible light. Improved ef-
“off-on” property for controllable release of drug [139]. It was realized ficacy of PDT was confirmed by in vitro assays, in which the as-obtained
by utilizing a ROS-sensitive thioketal linker to bind DOX and C60, which HA-TiO2-GO nanoplatform showed largely improved ability to kill
can be selectively broken by ROS. DOX was encapsulated efficiently by cancer cells than the HA-TiO2 or TiO2-GO complex. Furthermore, the
C60 at “off” state. Upon irradiation with a 532 nm laser, this nanosystem generated ROS can destroy GO, indicating the potential use this drug
was switched to “on” and C60 was excited to produce ROS to break the delivery system in clinical PDT in terms of the metabolism. In another
thioketal linker, leading to the precise release of DOX. This study re- example, Cho and Choi conjugated the PS Ce6 to GO via a redox-re-
vealed the potential application of C60-based nanoplatform in light- sponsive cleavable disulfide linker (GO-SS-Ce6), in which the fluores-
controlled drug delivery system to achieve tumor-targeting combined cence and the ROS generation were activated in the presence of in-
PDT/chemotherapy. In the work of Wang et al., C60 was doped with tracellular redox agents (such as glutathione) [154]. Without
mesoporous silica, forming a C60-silica nanoparticle system, which was glutathione, the fluorescence of Ce6 bound to GO was largely quenched
coated with hyaluronan for targeted therapy of cancer stem-like cells due to the FRET process, avoiding non-specific excitation and poor
(CSCs) [140]. By loading the chemotherapy drug DOX and photo- targeting abilities of PS. Compared to the free Ce6, GO-based Ce6
thermal agent indocyanine green (ICG), this C60-silica nanosystem was conjugate showed more efficient cellular internalization and pre-
found to be effective for the destruction of the breast CSCs through ferential accumulation of the PSs, and on-demand release of Ce6 inside
synergistic photodynamic/photothermal/chemo therapy under excita- A549 cancer cells. Additionally, the as-prepared GO-SS-Ce6 complex
tion at 800 nm. Li et al. reported the conjugation of graphene oxide was demonstrated to be an effective drug delivery vehicle, given the
(GO) to C60 to obtain a GO-C60 hybrid, which exhibited good stability high surface area and enhanced chemical tethering properties of GO.
under physiological conditions [141]. The introduction of GO can not In the study of Rong et al., GO was functionalized by PEG and was
only mediate the photothermal effects but also transmit the absorbed loaded with the PS 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-
light energy to C60 to trigger the ROS production of C60 under NIR alpha (HPPH) via supramolecular π-π stacking [155]. The radiolabeling
irradiation. As a consequence, the as-prepared GO-C60 hybrid can in- of HPPH with 64Cu allowed for in vivo fluorescence imaging and posi-
duce synergistic PDT/PTT effects on Hela cells in vitro. Very recently, tron emission tomography (PET) imaging, revealing that GO-PEG-
Iohara and co-workers used an anionic γ-cyclodextrin derivative, su- HPPH exhibited improved cellular uptake of the HPPH in comparison
gammadex, to largely improve the stability of C60 in physiological en- with free HPPH via a more active endocytosis strategy. As a result, GO-
vironment [142]. This was attributed to an electric layer generated by PEG-HPPH exhibited enhanced killing ability to 4T1 murine breast
the strong host-guest interactions between sugammadex and the surface cancer cells upon activation at 671 nm. Importantly, mice injected with
of C60. Moreover, the as-obtained sugammadex-C60 complex was tested GO-PEG-HPPH showed remarkably longer life time (~40 days) than the
by in vitro and in vivo experiments to show high antitumor ability ones under treatment with free HPPH (~24 days), indicating increased
through PDT effects. long-term survival of tumor-bearing mice after GO-PEG-HPPH treat-
ment. Similarly, Yan et al. loaded the PS sinoporphyrin sodium
3.6.3. Graphene-based nanomaterials (DVDMS) onto PEGylated GO, forming a novel phototheranostic na-
Due to the large surface area, outstanding thermal and optical noplatform GO-PEG-DVDMS [156]. The fluorescence intensity of
properties, as well as good biocompatibility [143–145], graphene-base DVDMS can be dramatically improved after loading of GO-PEG through
nanomaterials, including graphene quantum dots (GQDs), graphene intramolecular charge transfer, leading to enhanced fluorescence ima-
oxide (GO), and reduced GO (rGO), have been extensively exploited for ging. Furthermore, the GO-PEG nanoplatform can more effectively ac-
cancer therapy, such as anticancer drug delivery and PDT [146–148]. cumulate DVDMS in tumors through enhanced permeability and re-
GQDs have been demonstrated by Ge et al. to be a good PDT agent, tention (EPR) effect in comparison with free PS. As a consequence, PDT
which were able to give an excellent quantum yield of singlet oxygen studies revealed that complete destruction of the U87MG tumor in vivo
(up to 1.3) [149]. By employing the hydrothermal method, the GQDs was achieved after treatment with GO-PEG-DVDMS (Fig. 11). By in-
were synthesized using polythiophenes as the carbon precursor. The corporating with PEG-folate, GO has also been reported to be capable of
PDT effects of the GQDs were tested both in intro and in vivo, where the producing singlet oxygen intrinsically without the use of an organic PS
death of HeLa cells and the tumor decomposition in BALB/nu mice with [157]. The combined PDT/PTT effects mediated by the as-prepared GO-
breast cancer were observed. Unfortunately, the GQDs reported in this PEG-folate was demonstrated both in vitro and in vivo on the destruction
study showed photodynamic activity only when excited by the visible of B16F0 melanoma tumor with 980 nm light irradiation.
light. In 2018, Kuo et al. doped and functionalized the GQDs with ni- In addition to GO, rGO was also investigated for the theranostic use
trogen and an amino group, demonstrating that the corresponding in cancer. A nanosystem was reported by Zhang et al. for a combination
amino-N-GQDs showed outstanding singlet oxygen production ability of lysosome-targeted phosphorescent imaging and photothermal/pho-
in the NIR region (800 nm) [150]. Importantly, compared to the un- todynamic therapy, which was composed of rGO nanosheet and a PEG
modified GQDs, the as-prepared amino-N-GQDs turned out to be a more functionalized Ru(II) complex (Ru-PEG) [158]. The Ru-PEG complex,
efficient PDT agent with enhanced two-photon excitation. acting as PS and imaging agent, was decorated onto rGO via π-π
In comparison with graphene, GO exhibits more favorable proper- stacking and hydrophobic interactions. Under irradiation at 450 nm,
ties in terms of being a vehicle for delivering PSs, due to its improved poorer quantum yield of ROS produced by rGO-Ru-PEG was found than
water solubility and diverse functionalization chemistry. In addition, that by Ru-PEG (0.31 and 0.06, respectively), which was likely caused

11
J. Chen, et al. Biomaterials 237 (2020) 119827

Fig. 11. (a) Images of mice bearing U87MG tumor after treatment with different conditions. (b) Time-dependent changes of tumor volume in different groups.
Reproduced with permission [156]. Copyright 2015, Elsevier.

by the quenching effect of rGO. It was demonstrated that rGO was able but also offered some new benefits to further improve the therapeutic
to improve the cellular uptake of Ru-PEG by human lung cancer efficacy at the same time [161].
A549 cells with specific localization to lysosome. Dual-modal PTT and
PDT treatment by rGO-Ru-PEG exhibited higher anticancer efficacy as 3.7.1. Manganese dioxide (MnO2) nanosheets
compared with either treatment alone. Next to the generation of ROS, One of the issues for PDT-based tumor treatment is the consumption
cathepsin-initiated lysosomal damage by combined photothermal/ of generated ROS by the solid tumors that are mostly in a constant
photodynamic effect was also found to contribute to the apoptosis of hypoxic state. Moreover, the excessive expression of glutathione (GSH)
cancer cells. Moreover, rGO-Ru-PEG exhibited photothermal imaging in the tumor cells can consume the ROS produced by PDT agents as well
under illumination at 808 nm, which revealed the high antitumor ef- [186]. Therefore, the PDT efficacy and the clinical therapeutics of
ficacy in vivo by the prepared nanosystem. current PDT agents could be largely limited [187,188]. MnO2 na-
In short conclusion, numerous carbon-based nanomaterials have nosheets have been proven to be able to regenerate oxygen by reacting
been designed through surface modifications to increase the bio- with H2O2 within the tumor microenvironment, and decrease the
compatibility and tumor-targeting efficiency, exhibiting great potential amount of GSH from the reaction between them, thus rapidly increase
for PDT application. Also, it is feasible to control the structural and the oxygen level in the tumor tissues [161,189]. Besides, MnO2 na-
surface properties of carbon-based nanomaterials to get intended tox- nosheets also show some attractive features for use as a carrier for PSs.
icokinetics and pharmacokinetics [159]. Nevertheless, the biomedical For instance, they have strong PS adsorption ability due to electrostatic
use of carbon-based nanomaterials still faces a few problems, such as interaction and Mn–N coordinate bonds, which can promote their en-
the lack of standardized synthetic protocols for large-scale production docytosis in intracellular PDT [190]. Additionally, MnO2 nanosheets
with fixed properties, and dose-related toxicity in vivo [125,127]. For show good biocompatibility owing to the important role of nontoxic
instance, it has been documented that the lattice defects and oxygen- manganese in physiological metabolism [191]. From these perspec-
containing agents of CNTs may cause peroxidation damage, resulting in tives, extensive attentions have been attracted to investigate the use of
the toxicity [160]. MnO2 nanosheets in PDT.
In 2015, Shi group reported a MnO2 nanosheet-based theranostic
3.7. Two-dimensional (2D) nanomaterials nanosystem, where MnO2 nanosheets were used for immobilizing up-
conversion nanoprobes (UCSMs) loaded with PS silicon phthalocyanine
Since the first mechanical exfoliation of graphene was discovered in dihydroxide (SPCD) [192]. In the acidic tumor microenvironment,
2004, the development of 2D nanomaterials has achieved un- significantly increased signal intensity of oxygen was detected attrib-
precedented scientific and technological advances [161–163]. 2D na- uted to the redox reaction between MnO2 and endogenous H2O2, en-
nomaterials exhibit some unique physical and chemical features on hancing tumor saturated O2 by about 7%. Simultaneously, the
account of their large surface area, ultrathin thickness, tunable com- MnO2–H2O2 redox reaction also enhanced the upconversion lumines-
position and facile surface modifications [164–167]. These properties cence (UCL) emitted by UCSMs, which can be used for high-resolution
are highly desirable for diverse potential applications such as catalysis, tumor imaging (Fig. 12). In addition, singlet oxygen generated by the
device fabrication [168–170], optoelectronics [171–174], energy sto- PS-loaded UCSMs upon NIR irradiation was also improved by the O2
rage [175], sensing [176,177], as well as disease diagnosis [178–181] generation due to the UCSMs-H2O2 redox reaction, leading to increased
and therapeutics [182–185]. Therefore, 2D nanomaterials acting as PSs PDT efficacy in killing 4T1 murine breast cancer cells. MnO2 nanosheets
not only share the same advantages as the other types of nanomaterials, have also been used as a nanocarrier for PS Ce6, in which the MnO2

Fig. 12. Schematic illustration of the decomposition of MnO2 nanosheets from the UCSMs-H2O2 redox reaction, resulting in improved UCL imaging and oxygen
generation for enhancing the PDT effect. Reproduced with permission [192]. Copyright 2015, Wiley-VCH Verlag GmbH & Co. KGaA.

12
J. Chen, et al. Biomaterials 237 (2020) 119827

nanosheets also acted as an oxidant to consume the intracellular GSH in glycosheets onto n-type nitrogen doped reduced GO (n-rGO) through
order to achieve enhanced photodynamic efficiency [189]. In this na- p–n heterojunction, which was subsequently decorated with PEGylated-
nosystem, Ce6 was efficiently loaded on the MnO2 nanosheets and was modified MnO2 nanoparticles, forming a p-MoS2/n-rGO–MnO2–PEG
protected by MnO2 from self‐destruction under light exposure, so that nanocomposite [203]. In this nanosystem, the p–n heterojunction pro-
the PS can be effectively transported to cytoplasm. Remarkably, the as- motes the migration of electrons and holes to enhance the separation of
prepared Ce6–MnO2 nanocomplex outperformed the other nanoma- electron–hole pairs when exposed to NIR light [204], thus increasing
terial-based platforms (such as Ce6‐GO and Ce6‐MSN) in killing the ROS production through photocatalysis. In addition, MnO2 trig-
MCF‐7 cells under the conditions the authors used. The Ce6–MnO2- gered the disproportionation of intracellular H2O2 to generate more O2,
mediated PDT led to over 95% cell death, while these numbers for which in turn further improved the production of ROS. In vitro studies
Ce6‐GO and Ce6‐MSN nanosystems were 36% and 21%, respectively. revealed that this nanocomposite showed effective PDT effect in killing
These results demonstrated high potential of MnO2 nanosheets for im- HeLa cells. In 2019, a mesoporous silica nanorods (MSNR)/MoS2 na-
proving the PDT efficacy under a clinically relevant setting. nocomposite was constructed by Yang et al. [205], which was further
Zhang et al. deposited MnO2 nanosheets with TiO2-coated UCNPs, modified by human serum albumin (HSA) to improve the tumor tar-
which was further modified by amino-polyethylene glycol (PEG-NH2) geting ability and reduce the toxicity. Under excitation at 808 nm, MoS2
[193]. In addition to the oxygen generating from MnO2-catalyzed de- was able to exert photothermal effect that can lead to both the direct
composition of H2O2 for overcoming tumor hypoxia, the TiO2 nano- tumor destruction and the release of loaded PS Ce6 to generate ROS
shells can be activated by the UCL of UCNPs under NIR illumination to upon 660 nm light illumination, thereby achieving the synergetic
produced cytotoxic 1O2 and hydroxyl radicals (%OH) via water splitting, photothermal/photodynamic therapy. The anti-tumor effect has been
which are highly damaging to the tumor cells. Meanwhile, intracellular proven in both the in vitro experiments with breast cancer cells and in
superoxide dismutase (SOD) can catalyze the water-splitting process to vivo experiments with mice bearing 4T1 tumor.
produce by-product superoxide anion radicals (O2%-), thereby re- The nanomaterial-based combinational PTT/PDT treatment, which
plenishing more H2O2 and O2. In this sense, ROS generation was am- takes advantage of biomechanics and phototherapy, as well as the high
plified from reactive oxygen circulation, providing a significantly en- specific surface area of nanomaterials, has gained increasing popularity
hanced PDT outcome. Very recently, in another study by Zeng and co- in the field of multimodal therapy of cancer cells [195,206]. 2D TMDs,
authors, MnO2 nanosheets were modified with PEG-cyclic arginine- like WS2 nanosheets, have also been utilized in the combinational PTT/
glycineaspartic acid tripeptide (PEG-cRGD), which was subsequently PDT strategy. In 2014, Yong and co-authors fabricated WS2-based na-
conjugated with PS Ce6 with a high loading efficiency [194]. The nocomposites, in which PS MB was loaded on bovine serum albumin
prepared MnO2-PEG-cRGD/Ce6 nanocomplex can significantly elevate (BSA)-modified WS2 nanosheets [207]. The WS2 nanosheets were pre-
the oxygen concentration and was specifically uptaken by PC3 cells, pared through liquid exfoliation during which H2SO4 intercalation and
due to the cRGD-mediated tumor-targeted ability. As a result, favorable ultrasonication was adopted in order to improve the water and air
therapeutic outcomes were realized in vitro upon irradiation at 660 nm. stability. BSA was utilized to further functionalize the as-synthesized
WS2 nanosheets via physical adsorption to improve the biocompat-
3.7.2. 2D transition metal dichalcogenide (TMD) nanosheets ibility and dispersibility in physiological solutions, as well as the ad-
TMD nanosheets are a powerful tool for drug delivery and tumor sorption capability of loading the PS molecule thanks to BSA's amphi-
therapy, as they possess high specific surface area, good biocompat- philic molecular structure (Fig. 14). The as-obtained BSA–WS2@MB
ibility, facile modification, and ultrahigh light and heat conversion ef- nanocomplex exhibited photothermal effects under 808 nm irradiation,
ficiencies [195,196]. MoS2 nanosheets are one of the most re- leading to the release of MB molecules. Although the BSA–WS2@MB
presentative and explored 2D TMDs that attract a lot of research nanocomposites generated only little amount of singlet oxygen with
interests in the development and applications in biomedicine due to 665 nm LED lamp, the production of ROS was greatly improved under
several favorable features. First, the precursor for MoS2 nanosheets, NIR irradiation at 808 nm. The PTT and PDT dual synergistic therapy
molybdenite, is very abundant in nature [197]. Second, similar to other confirmed better therapeutic performance of the BSA–WS2@MB na-
2D nanomaterials, MoS2 nanosheets also represent many unique phy- nocomposites to kill the HeLa cancer cells, compared to the treatment
sical and chemical properties like large surface area and strong absor- using PTT or PDT alone. It was argued that the reason for enhanced
bance in NIR region [198,199]. Additionally, it is feasible to modify the PTT/PDT effects is that the NIR irradiation can promote both the
surface of MoS2 nanosheets, making them more promising for biome- photothermal effects of WS2 that resulted in accelerated release of PS
dical uses [200]. and the formation of singlet oxygen.
In 2014, Liu et al. used lipoic acid-terminated PEG (LA-PEG) to
modify MoS2 nanosheets, obtaining the PEGylated MoS2 (MoS2-PEG) 3.7.3. Graphitic-phase carbon nitride (g-C3N4) nanosheets
nanosystem that was highly stable in physiological solutions [201]. As one of the newly emerging 2D nanomaterials, g-C3N4 nanosheets
MoS2-PEG showed remarkably enhanced delivery efficacy of the PS Ce6 have been used in various biomedical applications, owing to their low
to cells, as much stronger intracellular Ce6 fluorescence was detected in toxicity, high stability, good biocompatibility, and high photo-
the cells incubated with MoS2-PEG/Ce6 in comparison with those in- luminescence quantum yields. The first example of their use in PDT was
cubated with free Ce6. In vitro experiments further demonstrated that reported by Lin et al. in 2014, in which the g-C3N4 nanosheets were
MoS2-PEG/Ce6 exhibited dramatically enhanced photodynamic ther- utilized as PS to generate singlet oxygen, achieving effective HeLa cells
apeutic efficacy in killing 4T1 cancer cells under 660 nm light exposure death upon light exposure with low power density (20 mW/cm2) at
compared to free Ce6, which was possibly caused by the promoted 440–450 nm [208]. Furthermore, by loading anticancer drug DOX, the
cellular uptake of the PS that led to more singlet oxygen produced in- g-C3N4 nanosheets can act as a carrier with high loading capacity of
side cells. In the work by Ji and co-workers [202], MoS2 nanosheets and 18,200 mg/g, owing to the high surface-to-volume ratio. The release of
fluorophore-labeled glycoligands could undergo self-assembly to form DOX was accelerated under acidic pH environment due to the promoted
the MoS2 glycosheets. The glycoligands can effectively and selectively protonation and solubility of DOX in the acidic condition. Additionally,
interact with the highly expressed galactose receptors on Hep-G2 cells, thanks to the high photoluminescence quantum yield of the g-C3N4
leading to the high tumor-specific efficiency of the as-obtained MoS2 nanosheets, it was possible to use them for imaging to track the delivery
glycosheets. Importantly, the MoS2 glycosheets have been proven to process.
show good singlet oxygen generation capability under light activation, In the work by Ma and co-workers, FeIII-modified g-C3N4 nanosheets
which eventually led to the death of Hep-G2 human liver cancer cells have been demonstrated to show outstanding catalytic ability of de-
(Fig. 13). More recently, Kapri and co-workers integrated p-type MoS2 composing H2O2 to produce O2 in cancer cells, thereby reducing tumor

13
J. Chen, et al. Biomaterials 237 (2020) 119827

Fig. 13. a) Structures of the glycoligands DK1, DK2,


and DK3 (Gal = galactose; GalNAc = N-acetyl ga-
lactosamine; Lac = lactose). b) Illustration of the
supramolecular self-assembly between MoS2 and DK,
and the targeted intracellular 1O2 generation via two
steps (I) receptor endocytosis and (II) light illumi-
nation. Reproduced with permission [202]. Copy-
right 2016, Wiley-VCH Verlag GmbH & Co. KGaA.

Fig. 14. Illustration of the preparation of BSA–WS2@MB nanocomposites and


their use as a PS delivery system for combinational PTT and PDT treatment.
Reproduced with permission [207]. Copyright 2014, The Royal Society of
Chemistry.

Fig. 15. Structure of PCCN and illustration of the enhancement of PDT by light-
hypoxia and enhancing the PDT outcome [209]. A mitochondria-tar- driven water splitting. Reproduced with permission [210]. Copyright 2016,
geting moiety, (4-carboxybutyl)triphenylphosphonium bromide (TPP), American Chemical Society.
was conjugated to deliver the C3N4–Fe nanosheets loaded with PS MB
to the mitochondria. Furthermore, the conjugation of TPP could also
photoexcitation of the g-C3N4 suspensions containing spherical gold
facilitate the sensitivity of cancer cells to produce singlet oxygen by
nanoparticles (Fig. 16) [211]. The incorporation of gold nanoparticles
improved mitochondria dysfunction, which further triggered the death
could improve the absorption efficiency of 670 nm light, hence sig-
of cells. As a result, nearly complete removal of mouse cervical tumor
nificantly improving the efficiency of 1O2 production through photo-
was achieved in vivo. Similarly, by doping with carbon dots, C3N4 na-
catalytic water-splitting process due to the enhanced electron/hole se-
nosheets have also shown the ability to generate O2 in tumor tissues
paration. Good PDT effects of the g-C3N4-AuNPs nanocomposites have
[210]. In this strategy, carbon dots have been utilized to enhance the
been demonstrated in the apoptosis and/or necrosis of three tested
red light absorption of the C3N4 nanosheets, which could induce in vivo
cancer cell lines (i.e., A549, MCF-7, and HeLa) upon 670 nm laser il-
water splitting under 630 nm light, thus increasing the intracellular O2
lumination. In addition, effective suppression of tumor growth has also
concentration. The PS protoporphyrin IX (PpIX) was linked with tumor-
been achieved in MCF-7 tumor-bearing mice.
targeting sequence RGD (Arg-Gly-Asp) via PEG, forming an amphi-
pathic polymer, which was subsequently assembled with carbon-dot-
doped C3N4 (CCN) via π-π stacking between PpIX and C3N4 to obtain 3.7.4. Black phosphorus (BP)
the PCCN nanoparticles (Fig. 15). Both in vitro and in vivo assays showed BP, as a new type of 2D semiconductor, has attracted extensive
that this as-obtained PCCN nanosystem showed superior ability over- scientific efforts in the field of PDT since the first successful exfoliation
come tumor hypoxia and enhanced PDT effects. Very recently, Dai et al. of bulk BP into single- or few-layer BP in 2014, owing to its exceptional
decorated the g-C3N4 nanosheets with gold nanoparticles via structural, optical and chemical properties [212–221]. First, the
bandgap of BP is tunable and is correlated to the number of layers, and

14
J. Chen, et al. Biomaterials 237 (2020) 119827

Fig. 16. Schematic illustration of the g-C3N4-AuNP-mediated PDT and the production of ROS under 670 nm irradiation. Reproduced with permission [211].
Copyright 2019, American Chemical Society.

it is feasible to obtain monolayer or few-layer BP nanosheets via ex- decorated with polyacrylic acid (PAA), which were subsequently in-
foliation [222]. This unique optical property, in combination with its corporated to the surface of BP nanosheets functionalized with PEG-
impressive carrier mobility, allows broad optical absorption of BP, NH2 by electrostatic interaction, giving the UCNPs-BPS nanosystem
which determines that it can be used effectively in many biomedical (Fig. 18a). Importantly, both in vitro and in vivo assays confirmed that
uses, including PDT, PTT and drug delivery [107,196,223–227]. the UCNPs-BPS composite with 808 nm light irradiation showed greater
Second, BP has ultrahigh surface-area-to-volume ratio owing to its ROS generation capability and improved effect of tumor inhibition in
single-atomic thickness, providing large amounts of anchoring points comparison with other groups under 650 nm and 980 nm light acti-
for anticancer drugs, which makes it an ideal candidate for nanocarrier vation (Fig. 18b–d), indicating the great potential of UCNPs-BPS as a
in drug delivery systems [228–230]. Most importantly, BP generally PDT agent. Yet, the relatively low quantum yield (< 3%) caused by the
exhibits better biocompatibility and biodegradability inside the human upconverted process for the energy donor still remained a problem for
body with respect to other nanomaterials. Phosphorus (P) is an essential its clinical application and industrialization [167]. To address this
element of the human body which accounts for about 1% of body issue, a novel BPs@Au@Fe3O4 nanocomposite was developed by as-
weight [228], indicating its high potential in biomedical applications. sembling iron oxide (Fe3O4) nanoparticles and gold nanoparticles on BP
Additionally, BP can be readily biodegraded to nontoxic phosphate, nanosheets through an ordinary electrostatic attraction method [236].
phosphite or other PxOy in the human body, thus exhibiting its safety With a combination of the PTT effect of gold nanoparticles, the tumor-
for in vivo cancer therapy [231–233]. targeting and MRI guiding abilities of Fe3O4 nanoparticles, as well as
The first use of BP nanosheets in PDT was documented by Wang and the PDT effect of BP nanosheets, this nanosystem showed enhanced
co-workers [234] in 2015. In this work, ultrathin BP nanosheets were therapeutic effect with improved targeting abilities. Furthermore, the
obtained from the exfoliation of bulk BP in water (Fig. 17a), with a mice bearing U14 tumor treated with BPs@Au@Fe3O4 upon 650 nm
thickness showing a height of about 2.0 nm (Fig. 17b). The as-obtained illumination showed significant suppression of tumor growth. This
BP nanosheets provided a high quantum yield of singlet oxygen (up to work revealed the new horizons of BP nanosheets for biomedicine.
0.91) upon irradiation with xenon lamp or 660 nm laser, demonstrating Very recently, in the work from Yang et al., polydopamine (PDA)
their attractive applications in PDT. In vitro cell viability tests revealed functionalized BP nanosheets were covalently bound to PS Ce6 and
significant death of human MDA-MB-231 breast cancer cells by the BP TPP, giving the BP@PDA–Ce6&TPP nanosystem [237]. In this nano-
nanosheets under 660 nm light illumination (Fig. 17c). Cell apoptosis platform, PDA coated BP showed strong absorption in the NIR region
test showed remarkable cell apoptosis of the cancer cells after treatment and Ce6 provided effective ROS formation. Furthermore, the TPP
of the BP nanosheets under irradiation (Fig. 17d). In vivo PDT studies moiety offered the mitochondria-targeting capacity. As a result, sig-
demonstrated high tumor-suppression effect of the BP nanosheets by nificantly enhanced killing ability of cancer cells and outstanding per-
intratumor injection of them in MDA-MB-231 breast tumor-bearing formance in tumor growth inhibition have been achieved in the vi-
mice combined with light irradiation (Fig. 17e). Typically, BP na- sualized synergistic therapy. In another study by Liu and co-workers
nosheets alone can only be activated by visible light to cause PDT ef- [238], Rhodamine B (RhB)-encapsulated MnO2 (R–MnO2) and fluor-
fects [228]. However, the use of irradiation wavelength within the escein isothiocyanate (FITC)-decorated BP with peptide modification
visible light region suffers the issue of low tissue penetration, which (FBP) were electrostatically assembled, forming a BP/MnO2 nanoplat-
limits its clinical therapeutics. To this end, a multifunctional composite form (Fig. 19). In this R–MnO2-FBP nanocomposite, R–MnO2 acts as the
in which BP nanosheets were integrated with UCNPs was reported by O2 supplier and indicator, which could induce the generation of O2
Lv et al. to achieve NIR-mediated PDT at 808 nm [235]. In this study, under the acidic and H2O2-rich condition upon targeted FA receptor
the energy donor, NaGd-F4:Yb,Er@Yb@Nd@Yb UCNPs were first (FR)-mediated endocytosis. Besides, the oxygen generation process can

15
J. Chen, et al. Biomaterials 237 (2020) 119827

Fig. 17. (a) Schematic illustration of the fabrication of ultrathin BP nanosheets


through water exfoliation of bulk BP. (b) Atomic force microscopy (AFM) image
of the prepared BP nanosheets and the corresponding height image. (c) Cell
Fig. 18. (a) Synthetic scheme of the preparation of the UCNPs-BPS composite.
viability with different concentrations of BP nanosheets. (d) Cell apoptosis
(b) Cancer cells with UCNPs-BPS upon irradiation with different wavelengths.
following transfection with PBS, BP nanosheets and BP nanosheets under light
(c) Photographs of tumor-bearing mice and tumors after different treatments for
irradiation. (e) Time-dependent tumor growth under different formulations,
14 days. (d) Changes of tumor volume and body weight in different groups.
and the corresponding photographs of tumors after the treatment. Reproduced
Reproduced with permission [235]. Copyright 2016, American Chemical So-
with permission [234]. Copyright 2015, American Chemical Society.
ciety.

be monitored by fluorescence and magnetic resonance (MR) imaging


attributed to the released dye RhB and Mn2+. The FBP moiety, acting as
the theranostic agent, could mediate highly efficient PDT both in vitro
and in vivo. Futhurmore, a self-feedback system can be built for the
therapeutic response by utilizing the activated caspase in the cell
apoptosis caused by oxygen-evolving PDT (Fig. 19).
In short summary, the past decade has witnessed unprecedented
advancements of 2D nanomaterials for the use as next-generation
technologies in PDT application, attributed to their unique structural
architecture, noteworthy physical and chemical characteristics, and
flexible surface chemistry. Still, several concerns need to be addressed
before practical biomedical use: (1) how to get appropriate lateral size
with large-scale production [239]; (2) how to improve the tumor-tar-
geting efficiency [239]; (3) how to enhance the biocompatibility and
reduce the toxicity of some 2D nanomaterials [240].

3.8. Other nanomaterials

3.8.1. Nanoscale metal-organic frameworks (MOFs)


Nanoscale MOFs (NMOFs) have emerged as a promising delivery
platform for PDT due to versatile functionalities of their chemical
Fig. 19. Schematic illustration of the fabrication of the R–MnO2-FBP nano-
compositions, the well-defined crystalline structures, high porosity, and platform and the uses for monitoring oxygen self-supply, enhancing PDT and
tunable framework stability [48,241–243]. In 2014, the Lin group de- evaluating the therapeutic effects. Reproduced with permission [238]. Copy-
signed an Hf−porphyrin NMOF consisting of a porous UiO NMOF right 2019, Elsevier.
structure and a H2DBP (5,15-di(p-benzoato)porphyrin) bridging ligand
[244]. The PS is well-isolated from the framework, therefore avoiding

16
J. Chen, et al. Biomaterials 237 (2020) 119827

the aggregation and self-quenching of PS. Furthermore, the promoted in KB-tumorbearing mice attributed to the folate-driven PDT.
intersystem crossing attributed to the coordination between the ligands
and heavy Hf centers, as well as the unique porous NMOF structure, 3.8.3. Transition metal carbides (TMCs)
enhanced the production efficiency and the diffusion ability of singlet Recently, TMCs, including Ti3C2 and W2C, have also been in-
oxygen. Consequently, strong photodynamic effects have been achieved vestigated considerably in the biomedical application, because of their
both in vitro cancer cell examination and in vivo tumor-bearing mice flexible surface functionalization, strong NIR light absorption ability
assays. Subsequently, the same group reduced to ligand H2DBP to and good biocompatibility [48,256]. Several studies have also reported
H2DBC (5,15-di(p-benzoato)-chlorin), thereby preparing the chlorin- their use in PDT. In 2017, Ti3C2 nanosheets have been prepared by Liu
based incorporated NMOF [245]. This DBC-UiO nanoplatform ex- et al. for the use in synergistic PTT/PDT/chemotherapy by using a
hibited a red (13 nm) shift and an increased extinction coefficient (11- surface modification method during the synthesis, resulting in a thin
fold) of the lowest-energy Q band compared to the previous DBP-UiO thickness of ca. 100 nm and high surface stability [257]. Generation of
structure. As a consequence, DBC-UiO outperformed DBP-UiO in the singlet oxygen has been observed under 808 nm excitation, presumably
terms of singlet oxygen generation efficiency and PDT performance on owing to the energy transfer of photoexcited electrons from Ti3C2 to
in vitro and in vivo tests. More recently, they incorporated the TBP oxygen. Additionally, by loading with the anti-cancer drug DOX and
(5,10,15,20-tetra(p-benzoato)porphyrin) ligand to Fe3O clusters, fabri- modifying with tumor-targeting agent hyaluronic acid (HA), this as-
cating the Fe-TBP nanoconstruct [246]. Particularly, the Fe3O clusters prepared multifunctional nanosystem exhibited tumor-specific accu-
were able to catalyze the decomposition of intracellular H2O2 to form mulation and effective ablation of human colon carcinoma tumor in
O2 via a Fenton-like reaction to overcome tumor hypoxia. Therefore, a vivo. In another work by Li and co-workers, W2C nanoparticles have
significantly enhanced PDT performance has been achieved for effec- been prepared from the generation of organic–inorganic nanostructures
tive inhibition of tumors. Similarly, He and co-authors have reported a triggered by ionic self-assembly [258]. By further functionalizing with
Mn-porphyrin based MOF consisting of the Mn-porphyrin ligands and HA, the W2C nanoparticles showed enhanced biocompatibility and
biocompatible Zr4+ ions, where Mn-porphyrin exhibited good cata- tumor-targeting efficiency. Upon irradiation with NIR II light
lytic ability to decompose H2O2 to O2, realizing the oxygen self-sup- (1064 nm), the as-obtained nanocomposite was able to produce both
plementing PDT [247]. Besides, the unique porous MOF frameworks hydroxyl radicals and singlet oxygen via type I and type II reaction
can prevent the self-aggregation of Mn–porphyrin and accelerate the mechanisms, respectively. In addition, the W2C nanoparticles also ex-
diffusion of H2O2 and generated O2. hibited photothermal activity upon 1064 excitation, thus allowing sy-
nergetic photodynamic/photothermal therapy for efficient eradication
3.8.2. Organic nano-agents of tumors.
Some recently developed nano-agents, such as nanostructured To sum up, thanks to the rapidly developed nanoscience in the field
phthalocyanine self-assemblies [248] and porphysome [249], have also of biomedicine over the last few decades, it is believed that an in-
been utilized for PDT application. In 2017, Li et al. reported the na- creasing number of new nanomaterials are expected to be emerged,
nostructured phthalocyanine self-assemblies prepared from zinc(II) bringing hope for offering promising alternative treatment modalities of
phthalocyanine building blocks decorated with triethylene glycol (TEG) cancer.
linkers as targeting agents [250]. The as-prepared nanoassemblies show
targeted protein-induced partial disassembly, due to the unique non- 4. Current challenges of photodynamic therapy
covalent interactions between the targeting agents and phthalocya-
nines. Hence, the nano-phthalocyanine assemblies possess switchable 4.1. Tumor targeting efficiency
photoactivity, which triggered the generation of ROS and enables
tumor-specific PDT. Subsequently, they have fabricated a nanos- The lack of good tumor targeting efficiency associated with most of
tructured assembly from a phthalocyanine derivative and chemother- traditional organic PSs has largely limited their clinical use. On one
apeutic drug mitoxantrone (MA) [251]. The molecular recognition in- hand, the reason for the higher concentration of some PSs in the tumor
teractions of these two building blocks are ideal for spontaneous tissue than the neighboring healthy tissues is still not clear, and this
assembly, therefore generating uniform nano-assemblies. This nano- tumor targeting ability is attributed to the unnormal physiological
composite underwent nucleic-acid-responsive ROS production and features of the tumors [4,13]. On the other hand, the inherent prop-
showed tumor targeting PDT in vivo. In addition, the pure phthalo- erties of PSs, such as the morphology, particle size, and surface mod-
cyanine molecule can also undergo self-assembly, which was able to ulation can dramatically influent the tumor targeting ability [48]. In
produce ROS through type I reaction [252]. The as-obtained nanos- this sense, improving the tumor targeting efficiency of the PSs could be
tructured phthalocyanine assemblies exhibited good PDT performance complicated and challengeable. To overcome this issue, targeting li-
on bacterial cells. By using albumin to trap the phthalocyanine mole- gands conjugated to PSs and surface modification of the PDT agents
cule, the nanostructured phthalocyanine self-assemblies with hydro- with targeting moieties could be adopted to deliver the photosensitizing
philic groups can exhibit disassembly [253]. As a result, time-modu- drugs specifically to the tumors (Table 2) [51,259–270]. The use of
lated PDT has been achieved via the albumin-driven production of ROS. nanomaterials in PDT will provide great potentials for surface func-
Very recently, Li and co-authors developed the size-controllable na- tionalization that can enhance the tumor targeting efficiency.
nostructured self-assemblies based on a silicon(IV) phthalocyanine-
biotin conjugate by using different amounts of surfactant Cremophor EL 4.2. Deep tissue PDT
[254]. The ROS generation ability was dependent on the particle size,
and decreased along with the increase of particle size. The nanoscale During the PDT with traditional earlier generation PSs, visible light
self-assemblies could undergo disassembly driven by a tumor-specific is normally required for photoexcitation. However, the penetration
biotin-binding protein, i.e., avidin, resulting in the PDT-based photo- depth of visible light is rather low (< 3 mm) because of strong light
cytotoxicity on HepG2 cells. Porphysomes present as a stable and effi- absorption in visible region by most tissue chromophores [271].
cient carrier for porphyrin. However, they were reported to covert the Therefore, the photodynamic outcome is significantly hampered by the
photodynamic activities to photothermal activities completely [255]. weakening light intensity along with the increase of tissue depth. One
By conjugating with a folate-receptor, the nanostructure of the folate- strategy to deal with this issue is to employ PSs that can be excited by
porphysomes was disrupted in cancer cells rapidly, hence reversing the NIR light, which is within the “optical window” of biological tissue. The
PTT effects back to PDT effects aroused by the densely loaded por- use of NIR light in PDT could not only offer deeper tumor therapy, but
phyrins [249]. Effective inhibition of tumor growth has been observed also decrease the phototoxicity to healthy tissues as compared to the

17
J. Chen, et al. Biomaterials 237 (2020) 119827

Table 2 independent modalities (such as PTT and chemotherapy) can also be


Some commonly used targeting conjugates for PDT. adopted to deal with tumor hypoxia [48,280].
Conjugate Targeted tumor/cancer cells Reference
4.4. More efficient and more reliable nanomaterial-based photosensitizers
Folic acid Brain, lungs and ovary tumors [259–262]
Low-density lipoprotein Colon, hepatoblastoma (HepG2) and [263,264]
Undoubtedly, the PS represents a pivotal role in PDT. Hence, the
(LDL) retinoblastoma tumors
Epidermal growth factor Breast carcinoma cells, epidermal [265,266]
PDT treatment results are largely related to its physical, chemical and
(EGF) carcinoma and ovarian cancer pharmacokinetic properties. The PSs with high ROS generation effi-
Retinoic acid Neuroblastoma SK-N-DZ cells [267] ciency, good biocompatibility and good stability under physiological
Monoclonal antibodies Ovarian and breast cancer cells [268] conditions, as well as high tumor targeting ability are still highly de-
(MAbs)
manded. This review highlights some of the recent studies on the ap-
Magnetoliposomes Melanoma [269]
Glucose and galactose K562 cells [270] plication of nanomaterials in PDT (Table 3). Each nanoconstruct pos-
Phenylalanine Transformed PAM212 keratinocyte [51] sesses certain fascinating features and clear advantages that can be
cells exploited for PDT. Yet, it should be realized that the defects of the
nanomaterials still exist, which limit their practical clinical use. For
instance, the toxicity associated with (heavy) metal-based nanomater-
ials, long multi-step preparation associated with UCNPs, and instability
associated with BP nanosheets, and many others as discussed above.
Therefore, extending the potentials of nanomaterials in PDT by devel-
oping more efficient and more reliable nanomaterials still needs ex-
tensive and unremitting efforts.

5. Conclusions and future perspectives

Throughout the over 100-year history of PDT, more than 1000


known PSs that are both natural and synthetic have been discovered
[70]. Yet, only a few of them have gained access into the clinical ap-
plications in the management of cancers, mainly because of the lim-
Fig. 20. Illustration of the use of UCNPs to convert low-energy NIR irradiation itations of conventional PSs [5]. It is still imperative to develop the
to high-energy UV/visible emission for activating the PS to produce ROS (ex- “ideal” PSs that can be excited efficiently by longer activation wave-
emplified by Type II reaction). lengths with higher yield of ROS [281–285], and that can be highly
selectively distributed to the targeted site [286–288]. Undoubtedly, the
road to reach this target and eventually to make PDT as a first-line
PDT with UV or visible light [48,272]. However, the shortcoming ac-
treatment strategy in treating cancers is still long and challengeable.
companied by the use of NIR light is the low energy that may not ef-
Nevertheless, the utilization of nanomaterial-based agents in PDT pre-
fectively trigger the PSs to produce sufficient ROS. Therefore, there are
sents a great stride forward towards this goal, which could greatly
increasing interests in using UCNPs for PDT. UCNPs could convert low-
tackle some of the shortcomings associated with conventional PSs. Next
energy NIR light, which has deep tissue penetration, to high-energy
to the exceptional physicochemical and optical properties of nanoma-
UV/visible light that could excite the accompanying PS to generate ROS
terials that make them more favorable to be used in PDT than the
(Fig. 20) [273]. The other strategy is to use the light source that are not
conventional PSs, the facile modifications and functionalization of the
restricted by tissue thickness, such as X-ray and internal lights. Never-
surface can offer additional versatility, such as the targeting moieties
theless, the low ROS generation efficiency and side effects to healthy
leading to improved specificity in the accumulation of PS-conjugated
tissue associated with X-ray and poor energy transfer to the PS asso-
nanomaterials at the target site [4]. In this review, the development of
ciated with internal lights indicate that more efforts are needed in these
various nano-agents that were reported for the application in PDT is
fields [48]. In general, to realize deep tissue PDT with high efficiency is
summarized.
still a challenging task.
In recent years, PDT-based multimodal treatment of cancer has
become a popular trend, in which PDT is carried out synergistically
4.3. Tumor hypoxia with other therapeutic methods, including PTT, MRI, drug delivery,
fluorescence imaging, chemotherapy and immunotherapy, etc. The
Apparently, the photodynamic effect is highly dependent on the combinational treatment can make use of the advantages of each
concentration of surrounding oxygen. Hence, the antitumor effects modality and compensate each's shortcomings, therefore representing a
elicited by PDT are significantly hampered in hypoxic tumors, in which more promising way to achieve safe and effective therapeutic results as
oxygen is largely consumed by vastly growing tumor cells. Besides, PDT compared to the single treatment strategy. Based on this principle, it is
is an oxygen consuming process that would aggravate the tumor hy- worth mentioning the combination of the smart NIR-responsive drug
poxia, thereby leading to decreased PDT efficiency. In order to deal delivery system and UCNP-based PDT, which shows some appealing
with tumor hypoxia, nanoplatforms consisting of PS and nanomaterial superiorities as a treatment modality of cancer. This multimodal system
that can catalyze the decomposition of H2O2 to produce O2, such as the utilizes the spatiotemporally controllable feature of NIR for on-demand
MnO2 nanosheets discussed above, were developed. Besides, some drug release, as well as the photon upconversion property of UCNPs to
techniques that can contribute to the reperfusion of O2, such as light generate high energy light for promoted drug release and effective ROS
fractionation for controlled “on” and “off” periods of light exposure production (Fig. 21) [289,290].
[274,275] and low fluence rate PDT [276–278], have also been used to Many other advanced techniques, such as X-ray light irradiation,
overcome tumor hypoxia. Unfortunately, these methods do not take microwave-induced PDT, bacteriophage nanowire- or stem cell-based
effect in the cases where tumor hypoxia is caused by rapid growth of PS delivery system, have also been used to improve the PDT effects
tumor cells [279]. Alternatively, the PSs that are independent of [291]. As mentioned earlier, X-ray possesses deep tissue penetration
oxygen, like TiO2 and g-C3N4, and PSs that can mediate the O2-in- capability [292,293], but also has weak PS activation ability and can
dependent type I reaction and combinational therapy with O2- lead to side effects to normal tissues. However, using heavy element-

18
J. Chen, et al.

Table 3
Summary of representative nanoplatforms for PDT.
Nanomaterial PS drug Modification agents Remarks Light source Applications Ref.

Au NRs - - Length: 37.3 ± 2.4 nm; 915 & 780 nm PDT/PTT for B16F0 melanoma tumor [62]
Diameters: 11 ± 1.2 nm
Au NEs – - Extinction coefficient: NIR I and NIR II PDT/PTT for B16F0 melanoma tumor [63]
~0.69 × 1012 M−1 cm−1 at 915 nm, 0.74 × 1012 M−1 cm−1 at
1064 nm
Silver nanoparticles - - Ag decahedrons (50 nm diameter) NIR PDT for HeLa cells [71]
Ag nanocubes (58 nm edge length)
Ag triangular plates (33 nm edge length, 7.5 nm thickness)
ORMOSIL nanoparticles IP - Size: ~20 nm; Highly monodispersed and stable in aqueous 514 nm PDT for RIF-1 tumor cells [76]
suspension
MSNs ZnPc PEG & PEI Diameter: 50 nm 680 nm PDT for H22 tumors [79]
Silica nanoparticles Ce6 FA Up to 100 μg/mL in MDA-MB-231 cells for 24 h 670 nm PDT for MDA-MB-231 cells [81]
Porous silicon nanoparticles – Gold nanoparticles & mannose 80 μg/mL in MCF‐7 breast cancer cells for 5h 800 nm Fluorescence imaging/PDT for MCF‐7 breast [87]
moieties cancer cells
1
CdTe QDs – TSPP O2 quantum yield: 0.43 355 nm Imaging and PDT [96]
1

19
NAC-CdTe/ZnS QDs R6G TMPyP & NIR775 O2 quantum yield: 0.91 400 nm Fluorescence imaging/PDT for KB tumors [98]
NaYF4:Yb/Er UCNPs ZnPc & MC-540 FA and PEG Size: ~80 nm 980 nm PDT for melanoma tumors [110]
NaGdF4 UCNPs Ce6 Gold nanorod dimer Size: 20 ± 2 nm PTT: 808 nm; PDT: PTT/PDT for HeLa tumors [123]
980 nm
SWCNTs DOX Fe3O4@CQDs & PEG Width: 35 nm 808 nm MRI/PTT/PDT for HeLa tumors [132]
C60 and mesoporous silica DOX Hyaluronan & ICG Diameter: ~60 nm 800 nm PTT/PDT/chemo therapy for breast CSCs [140]
N-GQDs – Amino group Lateral size: 7.24 ± 0.70 nm 800 nm Two-photon PDT for KB-50 cancer cells [150]
GO DVDMS PEG Size: < 50 nm; 630 nm Fluorescence imaging/PDT for U87MG tumors [156]
Thickness: ~2 nm
MnO2 nanosheets and UCSMs SPCD – 200 μg/mL in cells for 24 h NIR UCL imaging/PDT/RT for 4T1 tumors [192]
MoS2 nanosheets Ce6 LA-PEG Ce6 loading ratio: ~30% 660 nm PTT/PDT for 4T1 tumors [201]
WS2 nanosheets MB BSA Extinction coefficient: 21.8 L g−1 cm−1 808 nm PTT/PDT for HeLa tumors [207]
Diameter: 20–100 nm;
Thickness: 4–5 nm
Carbon-dot-doped C3N4 PpIX RGD & PEG Hydrodynamic size: 180 nm 630 nm O-elevated PDT for EGFP-4T1 tumors [210]
BP nanosheets – – Hight: 2.0 nm; 1O2 quantum yield: up to 0.91 660 nm PDT for MDA-MB-231 breast tumors [234]
BP nanosheets Ce6 PDA & TPP Thickness: 24.3 nm; Heigh: 49.1 nm; Biocompatibility: 25 μg/mL 660 nm PTT/PDT for tumors [237]
UiO NMOF – DBP Diameter: 76.3 nm; Biocompatibility: 30 μg/mL for 12 h 640 nm PDT for SQ20B tumors [244]
Nano-phthalocyanine assemblies – TEG Size: 100 nm 655 nm PDT for A549 tumors [250]
Ti3C2 nanosheets – Al(OH)4– Thickness: ~100 nm; 808 nm PTT/PDT/chemotherapy for HCT-116 tumors [257]
Extinction coefficient 28.6 Lg–1 cm−1
Biomaterials 237 (2020) 119827
J. Chen, et al. Biomaterials 237 (2020) 119827

Fig. 21. Illustration of multimodal cancer treatment with smart NIR-responsive drug delivery system and UCNP-based PDT. Reproduced with permission [289].
Copyright 2019, Elsevier.

Fig. 22. Some advanced techniques used for PDT. (a)


Schematic illustration of X-ray-induced PDT.
Reproduced with permission [296]. Copyright 2018,
The Royal Society of Chemistry. (b) Schematic illus-
tration of microwave-induced PDT. Reproduced with
permission [291]. Copyright 2019, Wiley-VCH Verlag
GmbH & Co. KGaA. (c) Schematic illustration of the
use of phages in PDT for delivering PS pyr-
opheophorbide a (PPa). Reproduced with permission
[305]. Copyright 2013, Wiley-VCH Verlag GmbH &
Co. KGaA. (d) Schematic illustration of the use of
mesenchymal stem cells (MSC) in PDT for delivering
PS-loaded nanoparticles. Pp-18 = purpurin-18. Re-
produced with permission [306]. Copyright 2014,
Wiley-VCH Verlag GmbH & Co. KGaA.

containing and X-ray sensitive nanoparticles, the photoelectric cross- developing new nanomaterial-based platforms for enhanced PDT and
sections can be largely increased, therefore reducing the radiation to the emerging new techniques for future enhancement of PDT, it is be-
healthy tissues [291]. In addition, these nanoparticles are able to lieved that PDT will gain acceptance as a potent therapeutic modality in
transfer the X-ray irradiation to light emission that can activate the PS clinical therapeutics of cancer.
to produce ROS (Fig. 22a) [292,294–299]. Microwave, on the other
hand, can not only induce local hyperthermia that makes tumors more
sensitive to chemotherapeutic drugs and ionizing radiation [300,301], Declaration of competing interest
but also trigger some nanoparticles to produce ROS [302]. Additionally,
the heat induced by microwave could increase the blood supply The authors declare that they have no conflict of interest.
through dilation of blood vessels, which in turn enhances the photo-
dynamic activity (Fig. 22b) [291]. In another advanced technique,
Acknowledgements
bacteriophage nanowires (phages) that show outstanding stability and
biocompatibility [303,304], are exploited for carrying photosensitizing
This work was partially supported by the Financial supports from
drugs (for instance, pyropheophorbide a) with high loading efficiency
the Science and Technology Development Fund (Nos. 007/2017/A1
via chemical conjugation [305]. Furthermore, the cancer-targeting
and 132/2017/A3), Macao Special Administration Region (SAR),
peptides attached to the phages can lead to selective destruction of
China, the State Key Research Development Program of China (Grant
SKBR-3 cancer cells (Fig. 22c) [305]. These results provided new stra-
No. 2019YFB2203503), National Natural Science Fund (Grant Nos.
tegies for PDT by developing various phage-based PS delivery systems.
61875138, U1801254, and 61961136001), and Science and
Lastly, stem cells that are naturally tumor-homing, are able to enhance
Technology Innovation Commission of Shenzhen
the accumulation of PSs in tumors when encapsulating PS-loaded na-
(KQTD2015032416270385, JCYJ20150625103619275, and
noparticles (Fig. 22d) [306,307]. The use of stem cells opens new in-
JCYJ20170811093453105). Authors also acknowledge the support
sights for further development of potent nanoplatforms for photo-
from Instrumental Analysis Center of Shenzhen University (Xili
dynamic cancer treatments. All in all, given the great achievements in
Campus).

20
J. Chen, et al. Biomaterials 237 (2020) 119827

References principles and clinical applications, Drug Discov. Today 4 (1999) 507–517,
https://doi.org/10.1016/S1359-6446(99)01412-9.
[27] X. Li, S. Lee, J. Yoon, Supramolecular photosensitizers rejuvenate photodynamic
[1] World Health Organization, Cancer, [Cited December 2018]. Available from: therapy, Chem. Soc. Rev. 47 (2018) 1174–1188, https://doi.org/10.1039/
https://www.who.int/en/news-room/fact-sheets/detail/cancer. c7cs00594f.
[2] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global [28] S. Zhu, R. Tian, A.L. Antaris, X. Chen, H. Dai, Near‐Infrared‐II molecular dyes for
cancer statistics 2018: GLOBOCAN estimates of incidence and mortality world- cancer imaging and surgery, Adv. Mater. (2019) 1900321, , https://doi.org/10.
wide for 36 cancers in 185 countries, CA, Canc. J. Clin. 68 (2018) 394–424, 1002/adma.201900321.
https://doi.org/10.3322/caac.21492. [29] G. Hong, A.L. Antaris, H. Dai, Near-infrared fluorophores for biomedical imaging,
[3] J. Zhang, C. Jiang, J.P. Figueiró Longo, R.B. Azevedo, H. Zhang, L.A. Muehlmann, Nat. Biomed. Eng. 1 (2017) 0010, , https://doi.org/10.1038/s41551-016-0010.
An updated overview on the development of new photosensitizers for anticancer [30] M. de Moraes, R.C. de Vasconcelos, J.P.F. Longo, L.A. Muehlmann, R.B. de
photodynamic therapy, Acta Pharm. Sin. B. 8 (2018) 137–146, https://doi.org/10. Azevedo, T.M.A.M. Lemos, A. de L.L. Costa, Effects of photodynamic therapy
1016/j.apsb.2017.09.003. mediated by nanoemulsion containing chloro-aluminum phthalocyanine: a histo-
[4] S.S. Lucky, K.C. Soo, Y. Zhang, Nanoparticles in photodynamic therapy, Chem. logic and immunohistochemical study in human gingiva, Photodiagnosis
Rev. 115 (2015) 1990–2042, https://doi.org/10.1021/cr5004198. Photodyn. Ther. 12 (2015) 592–597, https://doi.org/10.1016/J.PDPDT.2015.10.
[5] U. Chilakamarthi, L. Giribabu, Photodynamic therapy: past, present and future, 009.
Chem. Rec. 17 (2017) 775–802, https://doi.org/10.1002/tcr.201600121. [31] L.A. Muehlmann, M.C. Rodrigues, J.P.F. Longo, M.P. Garcia, K.R. Py-Daniel,
[6] L. Tran, C.T. Allen, R. Xiao, E. Moore, R. Davis, S.-J. Park, K. Spielbauer, C. Van A.B. Veloso, P.E.N. de Souza, S.W. da Silva, R.B. Azevedo, Aluminium-phthalo-
Waes, N.C. Schmitt, Cisplatin alters antitumor immunity and synergizes with PD- cyanine chloride nanoemulsions for anticancer photodynamic therapy: develop-
1/PD-L1 inhibition in head and neck squamous cell carcinoma, Canc. Immunol. ment and in vitro activity against monolayers and spheroids of human mammary
Res. 5 (2017) 1141–1151, https://doi.org/10.1158/2326-6066.CIR-17-0235. adenocarcinoma MCF-7 cells, J. Nanobiotechnol. 13 (2015) 36, https://doi.org/
[7] A. Ray, D.S. Das, Y. Song, T. Hideshima, Y.-T. Tai, D. Chauhan, K.C. Anderson, 10.1186/s12951-015-0095-3.
Combination of a novel HDAC6 inhibitor ACY-241 and anti-PD-L1 antibody en- [32] J.D. Miller, E.D. Baron, H. Scull, A. Hsia, J.C. Berlin, T. McCormick, V. Colussi,
hances anti-tumor immunity and cytotoxicity in multiple myeloma, Leukemia 32 M.E. Kenney, K.D. Cooper, N.L. Oleinick, Photodynamic therapy with the phtha-
(2018) 843–846, https://doi.org/10.1038/leu.2017.322. locyanine photosensitizer Pc 4: the case experience with preclinical mechanistic
[8] X. Gong, X. Li, T. Jiang, H. Xie, Z. Zhu, F. Zhou, C. Zhou, Combined radiotherapy and early clinical–translational studies, Toxicol. Appl. Pharmacol. 224 (2007)
and anti–PD-L1 antibody synergistically enhances antitumor effect in non–small 290–299, https://doi.org/10.1016/J.TAAP.2007.01.025.
cell lung cancer, J. Thorac. Oncol. 12 (2017) 1085–1097, https://doi.org/10. [33] A. Borgatti-Jeffreys, S.B. Hooser, M.A. Miller, R.M. Thomas, A. DeGortari,
1016/J.JTHO.2017.04.014. M.D. Lucroy, Preclinical evaluation of zinc phthalocyanine tetrasulfonate-based
[9] X. Hou, Y. Tao, Y. Pang, X. Li, G. Jiang, Y. Liu, Nanoparticle-based photothermal PDT, Proc. SPIE 5686 (2005) 624–630, https://doi.org/10.1117/12.588378.
and photodynamic immunotherapy for tumor treatment, Int. J. Canc. 143 (2018) [34] M. Wainwright, Photodynamic therapy: the development of new photosensitisers,
3050–3060, https://doi.org/10.1002/ijc.31717. anticancer, Agents Med. Chem. 8 (2008) 280–291, https://doi.org/10.2174/
[10] E.M.J. Diaz, E.M. Sturgis, G.E. Laramore, A.L. Sabichi, S.M. Lippman, G. Clayman, 187152008783961888.
Holland-frei Cancer Medicine, sixth ed., BC Decker, 2003, https://www.ncbi.nlm. [35] M.A. Biel, Photodynamic therapy of head and neck cancers, in: C. Gomer (Ed.),
nih.gov/books/NBK12354/. Methods Mol. Biol. Humana Press, Totowa, NJ, 2010, pp. 281–293, , https://doi.
[11] C. De Angelis, Side effects related to systemic cancer treatment: are we changing org/10.1007/978-1-60761-697-9_18.
the Promethean experience with molecularly targeted therapies? Curr. Oncol. 15 [36] E. Vogel, M. Köcher, H. Schmickler, J. Lex, Porphycene-a novel porphin isomer,
(2008) 198–199, https://doi.org/10.3747/co.v15i4.362. Angew Chem. Int. Ed. Engl. 25 (1986) 257–259, https://doi.org/10.1002/anie.
[12] H.L. Kaufman, J. Russell, O. Hamid, S. Bhatia, P. Terheyden, S.P. D'Angelo, 198602571.
K.C. Shih, C. Lebbé, G.P. Linette, M. Milella, I. Brownell, K.D. Lewis, J.H. Lorch, [37] M. Guardiano, R. Biolo, G. Jori, K. Schaffner, Tetra-n-propylporphycene as a tu-
K. Chin, L. Mahnke, A. von Heydebreck, J.M. Cuillerot, P. Nghiem, Avelumab in mour localizer: pharmacokinetic and phototherapeutic studies in mice, Canc. Lett.
patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a mul- 44 (1989) 1–6, https://doi.org/10.1016/0304-3835(89)90100-6.
ticentre, single-group, open-label, phase 2 trial, Lancet Oncol. 17 (2016) [38] G.M. Kitanov, Hypericin and pseudohypericin in some Hypericum species,
1374–1385, https://doi.org/10.1016/S1470-2045(16)30364-3. Biochem. Systemat. Ecol. 29 (2001) 171–178, https://doi.org/10.1016/S0305-
[13] T.J. Dougherty, C.J. Gomer, B.W. Henderson, G. Jori, D. Kessel, M. Korbelik, 1978(00)00032-6.
J. Moan, Q. Peng, Photodynamic therapy, J. Natl. Cancer Inst. 90 (1998) 889–905, [39] A. Kubin, F. Wierrani, U. Burner, G. Alth, W. Grunberger, Hypericin - the facts
https://doi.org/10.1093/jnci/90.12.889. about a controversial agent, Curr. Pharmaceut. Des. 11 (2005) 233–253, https://
[14] E.J. Hong, D.G. Choi, M.S. Shim, Targeted and effective photodynamic therapy for doi.org/10.2174/1381612053382287.
cancer using functionalized nanomaterials, Acta Pharm. Sin. B. 6 (2016) 297–307, [40] A.D. Garg, D.V. Krysko, P. Vandenabeele, P. Agostinis, Hypericin-based photo-
https://doi.org/10.1016/J.APSB.2016.01.007. dynamic therapy induces surface exposure of damage-associated molecular pat-
[15] D.E.J.G.J. Dolmans, D. Fukumura, R.K. Jain, Photodynamic therapy for cancer, terns like HSP70 and calreticulin, Cancer Immunol. Immunother. 61 (2012)
Nat. Rev. Canc. 3 (2003) 380–387, https://doi.org/10.1038/nrc1071. 215–221, https://doi.org/10.1007/s00262-011-1184-2.
[16] L. Brancaleon, H. Moseley, Laser and non-laser light sources for photodynamic [41] J.P. Tardivo, A. Del Giglio, C.S. de Oliveira, D.S. Gabrielli, H.C. Junqueira,
therapy, Laser Med. Sci. 17 (2002) 173–186, https://doi.org/10.1007/ D.B. Tada, D. Severino, R. de Fátima Turchiello, M.S. Baptista, Methylene blue in
s101030200027. photodynamic therapy: from basic mechanisms to clinical applications,
[17] T.J. Dougherty, G.B. Grindey, R. Fiel, K.R. Weishaupt, D.G. Boyle, Photoradiation Photodiagnosis Photodyn. Ther. 2 (2005) 175–191, https://doi.org/10.1016/
therapy. II. Cure of animal tumors with hematoporphyrin and light, JNCI J. Natl. S1572-1000(05)00097-9.
Canc. Inst. 55 (1975) 115–121, https://doi.org/10.1093/jnci/55.1.115. [42] T.B. Graciano, T.S. Coutinho, C.B. Cressoni, C. de P. Freitas, M.B.R. Pierre, S.A. de
[18] N. Yavari, S. Andersson-Engels, U. Segersten, P.-U. Malmstrom, An overview on Lima Pereira, M.M. Shimano, R. Cristina da Cunha Frange, M.T.J. Garcia, Using
preclinical and clinical experiences with photodynamic therapy for bladder chitosan gels as a toluidine blue O delivery system for photodynamic therapy of
cancer, Can. J. Urol. 18 (2011) 5778–5786 http://www.ncbi.nlm.nih.gov/ buccal cancer: in vitro and in vivo studies, Photodiagnosis Photodyn. Ther. 12
pubmed/21854709. (2015) 98–107, https://doi.org/10.1016/J.PDPDT.2014.11.003.
[19] B. Green, A.R.M. Cobb, C. Hopper, Photodynamic therapy in the management of [43] F. Harris, Z. Sayed, S. Hussain, D.A. Phoenix, An investigation into the potential of
lesions of the head and neck, Br. J. Oral Maxillofac. Surg. 51 (2013) 283–287, phenothiazinium-based photo-sensitisers to act as PDT agents, Photodiagnosis
https://doi.org/10.1016/J.BJOMS.2012.11.011. Photodyn. Ther. 1 (2004) 231–239, https://doi.org/10.1016/S1572-1000(04)
[20] K. Kostović, Z. Pastar, R. Ceović, Z.B. Mokos, D.S. Buzina, A. Stanimirović, 00046-8.
Photodynamic therapy in dermatology: current treatments and implications, Coll. [44] J. Atzpodien, S.C. Gulati, B.D. Clarkson, Comparison of the cytotoxic effects of
Antropol. 36 (2012) 1477–1481 http://www.ncbi.nlm.nih.gov/pubmed/ merocyanine-540 on leukemic cells and normal human bone marrow, Canc. Res.
23390855. 46 (1986) 4892–4895 http://www.ncbi.nlm.nih.gov/pubmed/3756850.
[21] N. Solban, I. Rizvi, T. Hasan, Targeted photodynamic therapy, Laser Surg. Med. 38 [45] E. Delaey, F. van Laar, D. De Vos, A. Kamuhabwa, P. Jacobs, P. de Witte, A
(2006) 522–531, https://doi.org/10.1002/lsm.20345. comparative study of the photosensitizing characteristics of some cyanine dyes, J.
[22] J. Krajczewski, K. Rucińska, H.E. Townley, A. Kudelski, Role of various nano- Photochem. Photobiol. B Biol. 55 (2000) 27–36, https://doi.org/10.1016/S1011-
particles in photodynamic therapy and detection methods of singlet oxygen, 1344(00)00021-X.
Photodiagnosis Photodyn. Ther. 26 (2019) 162–178, https://doi.org/10.1016/J. [46] E. Panzarini, V. Inguscio, G.M. Fimia, L. Dini, Rose bengal acetate PhotoDynamic
PDPDT.2019.03.016. therapy (RBAc-PDT) induces exposure and release of damage-associated molecular
[23] S. Schwartz, K. Absolon, H.V. Bull, Some relationships of porphyrins, X-rays and patterns (DAMPs) in human HeLa cells, PloS One 9 (2014) e105778, , https://doi.
tumors, Univ. Minn. Med. Bull. 27 (1955) 7–8. org/10.1371/journal.pone.0105778.
[24] M. Triesscheijn, P. Baas, J.H.M. Schellens, F.A. Stewart, Photodynamic therapy in [47] L.M. Moreira, F.V. Dos Santos, J.P. Lyon, M. Maftoum-Costa, C. Pacheco-Soares,
oncology, Oncol. 11 (2006) 1034–1044, https://doi.org/10.1634/theoncologist. N. Soares Da Silva, Photodynamic therapy: porphyrins and phthalocyanines as
11-9-1034. photosensitizers, Aust. J. Chem. 61 (2008) 741–754, https://doi.org/10.1071/
[25] D.A. Bellnier, W.R. Greco, G.M. Loewen, H. Nava, A.R. Oseroff, T.J. Dougherty, CH08145.
Clinical pharmacokinetics of the PDT photosensitizers porfimer sodium (photo- [48] M. Lan, S. Zhao, W. Liu, C. Lee, W. Zhang, P. Wang, Photosensitizers for photo-
frin), 2-[1-Hexyloxyethyl]-2-devinyl pyropheophorbide-a (photochlor) and 5-ALA- dynamic therapy, Adv. Health Mater. 8 (2019) 1900132, , https://doi.org/10.
induced protoporphyrin IX, Laser Surg. Med. 38 (2006) 439–444, https://doi.org/ 1002/adhm.201900132.
10.1002/lsm.20340. [49] D. Magda, R.A. Miller, Motexafin gadolinium: a novel redox active drug for cancer
[26] W.M. Sharman, C.M. Allen, J.E. van Lier, Photodynamic therapeutics: basic therapy, Semin. Canc. Biol. 16 (2006) 466–476, https://doi.org/10.1016/J.

21
J. Chen, et al. Biomaterials 237 (2020) 119827

SEMCANCER.2006.09.002. silica nanoparticle platforms for photodynamic therapy and their singlet oxygen
[50] S.W. Young, K.W. Woodburn, M. Wright, T.D. Mody, Q. Fan, J.L. Sessler, production and pH-dependent optical properties, Photochem. Photobiol. 78
W.C. Dow, R.A. Miller, Lutetium texaphyrin (PCI-0123): a near-infrared, water- (2007) 587–591, https://doi.org/10.1562/0031-8655(2003)0780587TEOMIS2.0.
soluble photosensitizer, photochem, Photobiol 63 (1996) 892–897, https://doi. CO2.
org/10.1111/j.1751-1097.1996.tb09647.x. [75] J. Lin, S. Wang, P. Huang, Z. Wang, S. Chen, G. Niu, W. Li, J. He, D. Cui, G. Lu,
[51] L. Bourré, F. Giuntini, I.M. Eggleston, M. Wilson, A.J. MacRobert, 5- X. Chen, Z. Nie, Photosensitizer-loaded gold vesicles with strong plasmonic cou-
Aminolaevulinic acid peptide prodrugs enhance photosensitization for photo- pling effect for imaging-guided photothermal/photodynamic therapy, ACS Nano 7
dynamic therapy, Mol. Canc. Therapeut. 7 (2008) 1720–1729, https://doi.org/10. (2013) 5320–5329, https://doi.org/10.1021/nn4011686.
1158/1535-7163.MCT-08-0092. [76] T.Y. Ohulchanskyy, I. Roy, L.N. Goswami, Y. Chen, E.J. Bergey, R.K. Pandey,
[52] P.P.S. Lee, P.C. Lo, E.Y.M. Chan, W.P. Fong, W.H. Ko, D.K.P. Ng, Synthesis and in A.R. Oseroff, P.N. Prasad, Organically modified silica nanoparticles with cova-
vitro photodynamic activity of novel galactose-containing phthalocyanines, lently incorporated photosensitizer for photodynamic therapy of cancer, Nano
Tetrahedron Lett. 46 (2005) 1551–1554, https://doi.org/10.1016/j.tetlet.2004. Lett. 7 (2007) 2835–2842, https://doi.org/10.1021/NL0714637.
12.137. [77] Y. Piao, A. Burns, J. Kim, U. Wiesner, T. Hyeon, Designed fabrication of silica-
[53] H. Kataoka, H. Nishie, N. Hayashi, M. Tanaka, A. Nomoto, S. Yano, T. Joh, New based nanostructured particle systems for nanomedicine applications, Adv. Funct.
photodynamic therapy with next-generation photosensitizers, Ann. Transl. Med. 5 Mater. 18 (2008) 3745–3758, https://doi.org/10.1002/adfm.200800731.
(2017) 183, https://doi.org/10.21037/14467. [78] P. Couleaud, V. Morosini, C. Frochot, S. Richeter, L. Raehm, J.-O. Durand, Silica-
[54] J. Taquet, C. Frochot, V. Manneville, M. Barberi-Heyob, Phthalocyanines cova- based nanoparticles for photodynamic therapy applications, Nanoscale 2 (2010)
lently bound to biomolecules for a targeted photodynamic therapy, Curr. Med. 1083–1095, https://doi.org/10.1039/c0nr00096e.
Chem. 14 (2007) 1673–1687, https://doi.org/10.2174/092986707780830970. [79] J. Tu, T. Wang, W. Shi, G. Wu, X. Tian, Y. Wang, D. Ge, L. Ren, Multifunctional
[55] B. Yang, Y. Chen, J. Shi, Reactive oxygen species (ROS)-Based nanomedicine, ZnPc-loaded mesoporous silica nanoparticles for enhancement of photodynamic
Chem. Rev. 119 (2019) 4881–4985, https://doi.org/10.1021/acs.chemrev. therapy efficacy by endolysosomal escape, Biomaterials 33 (2012) 7903–7914,
8b00626. https://doi.org/10.1016/J.BIOMATERIALS.2012.07.025.
[56] T. Kato, C.S. Jin, H. Ujiie, D. Lee, K. Fujino, H. Wada, H. Hu, R.A. Weersink, [80] M. Gary-Bobo, O. Hocine, D. Brevet, M. Maynadier, L. Raehm, S. Richeter,
J. Chen, M. Kaji, K. Kaga, Y. Matsui, B.C. Wilson, G. Zheng, K. Yasufuku, V. Charasson, B. Loock, A. Morère, P. Maillard, M. Garcia, J.-O. Durand, Cancer
Nanoparticle targeted folate receptor 1-enhanced photodynamic therapy for lung therapy improvement with mesoporous silica nanoparticles combining targeting,
cancer, Lung Canc. 113 (2017) 59–68, https://doi.org/10.1016/J.LUNGCAN. drug delivery and PDT, Int. J. Pharm. 423 (2012) 509–515, https://doi.org/10.
2017.09.002. 1016/J.IJPHARM.2011.11.045.
[57] W.-H. Tsai, K.-H. Yu, Y.-C. Huang, C.-I. Lee, EGFR-targeted photodynamic therapy [81] S. Bharathiraja, M.S. Moorthy, P. Manivasagan, H. Seo, K.D. Lee, J. Oh, Chlorin e6
by curcumin-encapsulated chitosan/TPP nanoparticles, Int. J. Nanomed. 13 conjugated silica nanoparticles for targeted and effective photodynamic therapy,
(2018) 903–916, https://doi.org/10.2147/IJN.S148305. Photodiagnosis Photodyn. Ther. 19 (2017) 212–220, https://doi.org/10.1016/j.
[58] S. Wang, R. Gao, F. Zhou, M. Selke, Nanomaterials and singlet oxygen photo- pdpdt.2017.06.001.
sensitizers: potential applications in photodynamic therapy, J. Mater. Chem. [82] C. Huang, Z. Zhang, Q. Guo, L. Zhang, F. Fan, Y. Qin, H. Wang, S. Zhou, W. Ou
(2004) 487–493, https://doi.org/10.1039/b311429e. Yang, H. Sun, X. Leng, X. Pan, D. Kong, L. Zhang, D. Zhu, A dual model imaging
[59] Christopher J. Ackerson, A. Pablo D. Jadzinsky, R.D. Kornberg, Thiolate ligands theragnostic system based on mesoporous silica nanoparticles for enhanced cancer
for synthesis of water-soluble gold clusters, J. Am. Chem. Soc. 127 (2005) phototherapy, Adv. Health Mater. 8 (2019) 1900840, https://doi.org/10.1002/
6550–6551, https://doi.org/10.1021/JA046114I. adhm.201900840.
[60] P.K. Jain, X. Huang, I.H. El-Sayed, M.A. El-Sayed, Noble metals on the nanoscale: [83] X. Li, H. Fan, T. Guo, H. Bai, N. Kwon, K.H. Kim, S. Yu, Y. Cho, H. Kim, K.T. Nam,
optical and photothermal properties and some applications in imaging, sensing, J. Yoon, X.-B. Zhang, W. Tan, Sequential protein-responsive nanophotosensitizer
biology, and medicine, Acc. Chem. Res. 41 (2008) 1578–1586, https://doi.org/10. complex for enhancing tumor-specific therapy, ACS Nano 13 (2019) 6702–6710,
1021/ar7002804. https://doi.org/10.1021/acsnano.9b01100.
[61] P. Ghosh, G. Han, M. De, C.K. Kim, Gold nanoparticles in delivery applications, [84] A.L. Lin, S.Z. Li, C.H. Xu, X.S. Li, B.Y. Zheng, J.J. Gu, M.R. Ke, J.D. Huang, A pH-
Adv. Drug Deliv. Rev. 60 (2008) 1307–1315, https://doi.org/10.1016/J.ADDR. responsive stellate mesoporous silica based nanophotosensitizer for: in vivo cancer
2008.03.016. diagnosis and targeted photodynamic therapy, Biomater. Sci. 7 (2019) 211–219,
[62] R. Vankayala, Y.-K. Huang, P. Kalluru, C.-S. Chiang, K.C. Hwang, First demon- https://doi.org/10.1039/c8bm00386f.
stration of gold nanorods-mediated photodynamic therapeutic destruction of tu- [85] M. Fujii, S. Minobe, M. Usui, S. Hayashi, E. Gross, J. Diener, D. Kovalev,
mors via near infra-red light activation, Small 10 (2014) 1612–1622, https://doi. Generation of singlet oxygen at room temperature mediated by energy transfer
org/10.1002/smll.201302719. from photoexcited porous Si, Phys. Rev. B Condens. Matter 70 (2004) 085311, ,
[63] P. Vijayaraghavan, C.-H. Liu, R. Vankayala, C.-S. Chiang, K.C. Hwang, Designing https://doi.org/10.1103/PhysRevB.70.085311.
multi-branched gold nanoechinus for NIR light activated dual modal photo- [86] L. Xiao, L. Gu, S.B. Howell, M.J. Sailor, Porous silicon nanoparticle photo-
dynamic and photothermal therapy in the second biological window, Adv. Mater. sensitizers for singlet oxygen and their phototoxicity against cancer cells, ACS
26 (2014) 6689–6695, https://doi.org/10.1002/adma.201400703. Nano 5 (2011) 3651–3659, https://doi.org/10.1021/nn1035262.
[64] R. Vankayala, C.-C. Lin, P. Kalluru, C.-S. Chiang, K.C. Hwang, Gold nanoshells- [87] A. Chaix, K. Rajoua, V. Stojanovic, K. El Cheikh, E. Bouffard, A. Brocéro,
mediated bimodal photodynamic and photothermal cancer treatment using ultra- M. Garcia, M. Maynadier, A. Morère, M. Gary-Bobo, F. Favier, J.-O. Durand,
low doses of near infra-red light, Biomaterials 35 (2014) 5527–5538, https://doi. F. Cunin, Two-photon fluorescence imaging and therapy of cancer cells with an-
org/10.1016/J.BIOMATERIALS.2014.03.065. isotropic gold-nanoparticle-supported porous silicon nanostructures, Chem. Nano.
[65] J. Lv, X. Zhang, N. Li, B. Wang, S. He, Absorption-dependent generation of singlet Mat. 4 (2018) 343–347, https://doi.org/10.1002/cnma.201700368.
oxygen from gold bipyramids excited under low power density, RSC Adv. 5 (2015) [88] Q. Jia, M. Chen, Q. Liu, W. Liu, H. Zhang, J. Ge, P. Wang, Ethylene glycol-medi-
81897–81904, https://doi.org/10.1039/C5RA15362J. ated synthetic route for production of luminescent silicon nanorod as photo-
[66] R. Vankayala, A. Sagadevan, P. Vijayaraghavan, C.-L. Kuo, K.C. Hwang, Metal dynamic therapy agent, Sci. China Mater. 60 (2017) 881–891, https://doi.org/10.
nanoparticles sensitize the formation of singlet oxygen, Angew. Chem. Int. Ed. 50 1007/s40843-017-9101-5.
(2011) 10640–10644, https://doi.org/10.1002/anie.201105236. [89] C. Zhang, L.W. Johnson, Quantifying RNA−Peptide interaction by single-quantum
[67] G. Pasparakis, Light-induced generation of singlet oxygen by naked gold nano- dot-based nanosensor: an approach for drug screening, Anal. Chem. 79 (2007)
particles and its implications to cancer cell phototherapy, Small 9 (2013) 7775–7781, https://doi.org/10.1021/AC071225W.
4130–4134, https://doi.org/10.1002/smll.201301365. [90] A.C.S. Samia, S. Dayal, C. Burda, Quantum dot-based energy transfer: perspectives
[68] C. Jiang, T. Zhao, P. Yuan, N. Gao, Y. Pan, Z. Guan, N. Zhou, Q.-H. Xu, Two-photon and potential for applications in photodynamic therapy, Photochem. Photobiol. 82
induced photoluminescence and singlet oxygen generation from aggregated gold (2006) 617–625, https://doi.org/10.1562/2005-05-11-IR-525.
nanoparticles, ACS Appl. Mater. Interfaces 5 (2013) 4972–4977, https://doi.org/ [91] A.R. Clapp, T. Pons, I.L. Medintz, J.B. Delehanty, J.S. Melinger, T. Tiefenbrunn,
10.1021/am4007403. P.E. Dawson, B.R. Fisher, B. O'Rourke, H. Mattoussi, Two-photon excitation of
[69] K. Sztandera, M. Gorzkiewicz, B. Klajnert-Maculewicz, Gold nanoparticles in quantum-dot-based fluorescence resonance energy transfer and its applications,
cancer treatment, Mol. Pharm. 16 (2019) 1–23, https://doi.org/10.1021/acs. Adv. Mater. 19 (2007) 1921–1926, https://doi.org/10.1002/adma.200602036.
molpharmaceut.8b00810. [92] S. Dayal, C. Burda, Semiconductor quantum dots as two-photon sensitizers, J. Am.
[70] J. Sun, S. Kormakov, Y. Liu, Y. Huang, D. Wu, Z. Yang, Recent progress in metal- Chem. Soc. 130 (2008) 2890–2891, https://doi.org/10.1021/JA0781285.
based nanoparticles mediated photodynamic therapy, Molecules 23 (2018) 1704, [93] D. Zhou, J.D. Piper, C. Abell, D. Klenerman, D.-J. Kang, L. Ying, Fluorescence
https://doi.org/10.3390/molecules23071704. resonance energy transfer between a quantum dot donor and a dye acceptor at-
[71] R. Vankayala, C.-L. Kuo, A. Sagadevan, P.-H. Chen, C.-S. Chiang, K.C. Hwang, tached to DNA, Chem. Commun. (2005) 4807–4809, https://doi.org/10.1039/
Morphology dependent photosensitization and formation of singlet oxygen (1Δg) b508911e.
by gold and silver nanoparticles and its application in cancer treatment, J. Mater. [94] Z.-D. Qi, D.-W. Li, P. Jiang, F.-L. Jiang, Y.-S. Li, Y. Liu, W.-K. Wong, K.-W. Cheah,
Chem. B. 1 (2013) 4379, https://doi.org/10.1039/c3tb20806k. Biocompatible CdSe quantum dot-based photosensitizer under two-photon ex-
[72] A. Sharma, A.K. Goyal, G. Rath, Recent advances in metal nanoparticles in cancer citation for photodynamic therapy, J. Mater. Chem. 9 (2011) 2455–2458, https://
therapy, J. Drug Target. 26 (2018) 617–632, https://doi.org/10.1080/1061186X. doi.org/10.1039/c0jm03229h.
2017.1400553. [95] I.L. Medintz, A.R. Clapp, H. Mattoussi, E.R. Goldman, B. Fisher, J.M. Mauro, Self-
[73] Y. Wang, F. Yang, H.X. Zhang, X.Y. Zi, X.H. Pan, F. Chen, W.D. Luo, J.X. Li, assembled nanoscale biosensors based on quantum dot FRET donors, Nat. Mater. 2
H.Y. Zhu, Y.P. Hu, Cuprous oxide nanoparticles inhibit the growth and metastasis (2003) 630–638, https://doi.org/10.1038/nmat961.
of melanoma by targeting mitochondria, Cell Death Dis. 4 (2013) e783, https:// [96] L. Shi, B. Hernandez, M. Selke, Singlet oxygen generation from water-soluble
doi.org/10.1038/cddis.2013.314. quantum dot-organic dye nanocomposites, J. Am. Chem. Soc. 128 (2006)
[74] F. Yan, R. Kopelman, The embedding of meta-tetra(hydroxyphenyl)-chlorin into 6278–6279, https://doi.org/10.1021/JA057959C.

22
J. Chen, et al. Biomaterials 237 (2020) 119827

[97] I. V Martynenko, V.A. Kuznetsova, А.O. Orlova, P.A. Kanaev, V.G. Maslov, P. Schlag, Hyperthermia in combined treatment of cancer, Lancet Oncol. 3 (2002)
A. Loudon, V. Zaharov, P. Parfenov, Y.K. Gun’ko, A. V Baranov, A. V Fedorov, 487–497, https://doi.org/10.1016/S1470-2045(02)00818-5.
Chlorin e6–ZnSe/ZnS quantum dots based system as reagent for photodynamic [123] M. Sun, L. Xu, W. Ma, X. Wu, H. Kuang, L. Wang, C. Xu, Hierarchical plasmonic
therapy, Nanotechnology 26 (2015) 055102, , https://doi.org/10.1088/0957- nanorods and upconversion core-satellite nanoassemblies for multimodal imaging-
4484/26/5/055102. guided combination phototherapy, Adv. Mater. 28 (2016) 898–904, https://doi.
[98] Y. Shen, Y. Sun, R. Yan, E. Chen, H. Wang, D. Ye, J.-J. Xu, H.-Y. Chen, Rational org/10.1002/adma.201505023.
engineering of semiconductor QDs enabling remarkable 1O2 production for [124] Y. Liu, X. Meng, W. Bu, Upconversion-based photodynamic cancer therapy, Coord.
tumor-targeted photodynamic therapy, Biomaterials 148 (2017) 31–40, https:// Chem. Rev. 379 (2019) 82–98, https://doi.org/10.1016/J.CCR.2017.09.006.
doi.org/10.1016/J.BIOMATERIALS.2017.09.026. [125] J. Saleem, L. Wang, C. Chen, Carbon-based nanomaterials for cancer therapy via
[99] Y. Zhang, G. Hong, Y. Zhang, G. Chen, F. Li, H. Dai, Q. Wang, Ag2S quantum dot: a targeting tumor microenvironment, Adv. Health Mater. 7 (2018) 1800525, ,
bright and biocompatible fluorescent nanoprobe in the second near-infrared https://doi.org/10.1002/adhm.201800525.
window, ACS Nano 6 (2012) 3695–3702, https://doi.org/10.1021/nn301218z. [126] D. Maiti, X. Tong, X. Mou, K. Yang, Carbon-based nanomaterials for biomedical
[100] C.-Y. Hsu, C.-W. Chen, H.-P. Yu, Y.-F. Lin, P.-S. Lai, Bioluminescence resonance applications: a recent study, Front. Pharmacol. 9 (2019) 1401, https://doi.org/10.
energy transfer using luciferase-immobilized quantum dots for self-illuminated 3389/fphar.2018.01401.
photodynamic therapy, Biomaterials 34 (2013) 1204–1212, https://doi.org/10. [127] D. Lu, R. Tao, Z. Wang, Carbon-based materials for photodynamic therapy: a mini-
1016/J.BIOMATERIALS.2012.08.044. review, Front. Chem. Sci. Eng. 13 (2019) 310–323, https://doi.org/10.1007/
[101] Y. Liu, Q. Yu, J. Chang, C. Wu, Nanobiomaterials: from 0D to 3D for tumor therapy s11705-018-1750-7.
and tissue regeneration, Nanoscale 11 (2019) 13678–13708, https://doi.org/10. [128] A. Battigelli, C. Ménard-Moyon, A. Bianco, Carbon nanomaterials as new tools for
1039/C9NR02955A. immunotherapeutic applications, J. Mater. Chem. B. 2 (2014) 6144–6156, https://
[102] W. Kuzyniak, O. Adegoke, K. Sekhosana, S. D'Souza, S.C. Tshangana, B. Hoffmann, doi.org/10.1039/C4TB00563E.
E.A. Ermilov, T. Nyokong, M. Höpfner, Synthesis and characterization of quantum [129] T. Murakami, H. Nakatsuji, M. Inada, Y. Matoba, T. Umeyama, M. Tsujimoto,
dots designed for biomedical use, Int. J. Pharm. 466 (2014) 382–389, https://doi. S. Isoda, M. Hashida, H. Imahori, Photodynamic and photothermal effects of
org/10.1016/j.ijpharm.2014.03.037. semiconducting and metallic-enriched single-walled carbon nanotubes, J. Am.
[103] N. Chen, Y. He, Y. Su, X. Li, Q. Huang, H. Wang, X. Zhang, R. Tai, C. Fan, The Chem. Soc. 134 (2012) 17862–17865, https://doi.org/10.1021/ja3079972.
cytotoxicity of cadmium-based quantum dots, Biomaterials 33 (2012) 1238–1244, [130] L. Wang, J. Shi, R. Liu, Y. Liu, J. Zhang, X. Yu, J. Gao, C. Zhang, Z. Zhang,
https://doi.org/10.1016/j.biomaterials.2011.10.070. Photodynamic effect of functionalized single-walled carbon nanotubes: a potential
[104] J. Yao, P. Li, L. Li, M. Yang, Biochemistry and biomedicine of quantum dots: from sensitizer for photodynamic therapy, Nanoscale 6 (2014) 4642–4651, https://doi.
biodetection to bioimaging, drug discovery, diagnostics, and therapy, Acta org/10.1039/C3NR06835H.
Biomater. 74 (2018) 36–55, https://doi.org/10.1016/j.actbio.2018.05.004. [131] A. Staicu, A. Smarandache, A. Pascu, M.L. Pascu, Photophysics of covalently
[105] J. Zhou, Z. Liu, F. Li, Upconversion nanophosphors for small-animal imaging, functionalized single wall carbon nanotubes with verteporfin, Appl. Surf. Sci. 417
Chem. Soc. Rev. 41 (2012) 1323–1349, https://doi.org/10.1039/C1CS15187H. (2017) 170–174, https://doi.org/10.1016/J.APSUSC.2017.03.031.
[106] X. Liu, C.-H. Yan, J.A. Capobianco, Photon upconversion nanomaterials, Chem. [132] M. Zhang, W. Wang, Y. Cui, X. Chu, B. Sun, N. Zhou, J. Shen, Magnetofluorescent
Soc. Rev. 44 (2015) 1299–1301, https://doi.org/10.1039/C5CS90009C. Fe3O4/carbon quantum dots coated single-walled carbon nanotubes as dual-
[107] H. Qiu, M. Tan, T. Ohulchanskyy, J. Lovell, G. Chen, Recent progress in upcon- modal targeted imaging and chemo/photodynamic/photothermal triple-modal
version photodynamic therapy, Nanomaterials 8 (2018) 344, https://doi.org/10. therapeutic agents, Chem. Eng. J. 338 (2018) 526–538, https://doi.org/10.1016/
3390/nano8050344. J.CEJ.2018.01.081.
[108] P. Zhang, W. Steelant, M. Kumar, M. Scholfield, Versatile photosensitizers for [133] C.-M. Lin, T.-Y. Lu, C60 fullerene derivatized nanoparticles and their application
photodynamic therapy at infrared excitation, J. Am. Chem. Soc. 129 (2007) to therapeutics, Recent Pat. Nanotechnol. 6 (2012) 105–113, https://doi.org/10.
4526–4527, https://doi.org/10.1021/JA0700707. 2174/187221012800270135.
[109] D.P. Valenzeno, J. Trudgen, A. Hutzenbuhler, M. Milne, Singlet oxygen involve- [134] Q. Li, Y. Xiu, X. Zhang, R. Liu, Q. Du, X. Shun, S. Chen, W. Li, Preparation of 99
ment in photohemolysis sensitized by merocyanine‐540 and rose bengal, mTc-C60(OH)x and its biodistribution studies, Nucl. Med. Biol. 29 (2002)
Photochem. Photobiol. 46 (1987) 985–990, https://doi.org/10.1111/j.1751-1097. 707–710, https://doi.org/10.1016/S0969-8051(02)00313-X.
1987.tb04881.x. [135] J. Wang, C. Chen, B. Li, H. Yu, Y. Zhao, J. Sun, Y. Li, G. Xing, H. Yuan, J. Tang,
[110] N.M. Idris, M.K. Gnanasammandhan, J. Zhang, P.C. Ho, R. Mahendran, Y. Zhang, Z. Chen, H. Meng, Y. Gao, C. Ye, Z. Chai, C. Zhu, B. Ma, X. Fang, L. Wan,
In vivo photodynamic therapy using upconversion nanoparticles as remote-con- Antioxidative function and biodistribution of [Gd@C82(OH)22]n nanoparticles in
trolled nanotransducers, Nat. Med. 18 (2012) 1580–1585, https://doi.org/10. tumor-bearing mice, Biochem. Pharmacol. 71 (2006) 872–881, https://doi.org/
1038/nm.2933. 10.1016/J.BCP.2005.12.001.
[111] C. Wang, H. Tao, L. Cheng, Z. Liu, Near-infrared light induced in vivo photo- [136] L.W. Zhang, J. Yang, A.R. Barron, N.A. Monteiro-Riviere, Endocytic mechanisms
dynamic therapy of cancer based on upconversion nanoparticles, Biomaterials 32 and toxicity of a functionalized fullerene in human cells, Toxicol. Lett. 191 (2009)
(2011) 6145–6154, https://doi.org/10.1016/J.BIOMATERIALS.2011.05.007. 149–157, https://doi.org/10.1016/J.TOXLET.2009.08.017.
[112] F. Chen, S. Zhang, W. Bu, Y. Chen, Q. Xiao, J. Liu, H. Xing, L. Zhou, W. Peng, [137] D.W. Cagle, S.J. Kennel, S. Mirzadeh, J.M. Alford, L.J. Wilson, In vivo studies of
J. Shi, A uniform sub-50 nm-sized magnetic/upconversion fluorescent bimodal fullerene-based materials using endohedral metallofullerene radiotracers, Proc.
imaging agent capable of generating singlet oxygen by using a 980 nm laser, Natl. Acad. Sci. U. S. A. 96 (1999) 5182–5187, https://doi.org/10.1073/pnas.96.
Chem. Eur J. 18 (2012) 7082–7090, https://doi.org/10.1002/chem.201103611. 9.5182.
[113] B. del Rosal, D. Jaque, Upconversion nanoparticles for in vivo applications: lim- [138] Z. Li, F. Zhang, L. Pan, X. Zhu, Z. Zhang, Preparation and characterization of in-
itations and future perspectives, Methods Appl. Fluoresc. 7 (2019) 022001, , jectable Mitoxantrone poly (lactic acid)/fullerene implants for in vivo chemo-
https://doi.org/10.1088/2050-6120/ab029f. photodynamic therapy, J. Photochem. Photobiol. B Biol. 149 (2015) 51–57,
[114] M.-H. Chan, Y.-T. Pan, I.-J. Lee, C.-W. Chen, Y.-C. Chan, M. Hsiao, F. Wang, L. Sun, https://doi.org/10.1016/J.JPHOTOBIOL.2015.05.018.
X. Chen, R.-S. Liu, Minimizing the heat effect of photodynamic therapy based on [139] J. Shi, B. Wang, L. Wang, T. Lu, Y. Fu, H. Zhang, Z. Zhang, Fullerene (C60)-based
inorganic nanocomposites mediated by 808 nm near-infrared light, Small 13 tumor-targeting nanoparticles with “off-on” state for enhanced treatment of
(2017) 1700038, https://doi.org/10.1002/smll.201700038. cancer, J. Contr. Release 235 (2016) 245–258, https://doi.org/10.1016/J.
[115] X.J. Loh, Q.Q. Dou, E. Ye, C.P. Teng, Effective near-infrared photodynamic therapy JCONREL.2016.06.010.
assisted by upconversion nanoparticles conjugated with photosensitizers, Int. J. [140] H. Wang, P. Agarwal, S. Zhao, J. Yu, X. Lu, X. He, Combined cancer therapy with
Nanomed. 10 (2015) 419–432, https://doi.org/10.2147/IJN.S74891. hyaluronan-decorated fullerene-silica multifunctional nanoparticles to target
[116] Q. Yu, E.M. Rodriguez, R. Naccache, P. Forgione, G. Lamoureux, F. Sanz- cancer stem-like cells, Biomaterials 97 (2016) 62–73, https://doi.org/10.1016/J.
Rodriguez, D. Scheglmann, J.A. Capobianco, Chemical modification of temoporfin BIOMATERIALS.2016.04.030.
– a second generation photosensitizer activated using upconverting nanoparticles [141] Q. Li, L. Hong, H. Li, C. Liu, Graphene oxide-fullerene C60 (GO-C60) hybrid for
for singlet oxygen generation, Chem. Commun. 50 (2014) 12150–12153, https:// photodynamic and photothermal therapy triggered by near-infrared light, Biosens.
doi.org/10.1039/C4CC05867D. Bioelectron. 89 (2017) 477–482, https://doi.org/10.1016/J.BIOS.2016.03.072.
[117] S. Gai, G. Yang, P. Yang, F. He, J. Lin, D. Jin, B. Xing, Recent advances in func- [142] D. Iohara, F. Hirayama, M. Anraku, K. Uekama, Physiologically stable hydrophilic
tional nanomaterials for light–triggered cancer therapy, Nano Today 19 (2018) C 60 nanoparticles for photodynamic therapy, ACS Appl. Nano Mater. 2 (2019)
146–187, https://doi.org/10.1016/J.NANTOD.2018.02.010. 716–725, https://doi.org/10.1021/acsanm.8b01862.
[118] W. Fan, W. Bu, J. Shi, On the latest three-stage development of nanomedicines [143] Y.-W. Chen, Y.-L. Su, S.-H. Hu, S.-Y. Chen, Functionalized graphene nanocompo-
based on upconversion nanoparticles, Adv. Mater. 28 (2016) 3987–4011, https:// sites for enhancing photothermal therapy in tumor treatment, Adv. Drug Deliv.
doi.org/10.1002/adma.201505678. Rev. 105 (2016) 190–204, https://doi.org/10.1016/J.ADDR.2016.05.022.
[119] B. del Rosal, B. Jia, D. Jaque, Beyond phototherapy: recent advances in multi- [144] L. Yi, Y. Zhang, X. Shi, X. Du, X. Wang, A. Yu, G. Zhai, Recent progress of func-
functional fluorescent nanoparticles for light-triggered tumor theranostics, Adv. tionalised graphene oxide in cancer therapy, J. Drug Target. 27 (2019) 125–144,
Funct. Mater. 28 (2018) 1803733, , https://doi.org/10.1002/adfm.201803733. https://doi.org/10.1080/1061186X.2018.1474359.
[120] J. Zuo, L. Tu, Q. Li, Y. Feng, I. Que, Y. Zhang, X. Liu, B. Xue, L.J. Cruz, Y. Chang, [145] C. Xing, G. Jing, X. Liang, M. Qiu, Z. Li, R. Cao, X. Li, D. Fan, H. Zhang, Graphene
H. Zhang, X. Kong, Near infrared light sensitive ultraviolet–blue nanophotoswitch oxide/black phosphorus nanoflake aerogels with robust thermo-stability and sig-
for imaging-guided “off–on” therapy, ACS Nano 12 (2018) 3217–3225, https:// nificantly enhanced photothermal properties in air, Nanoscale 9 (2017)
doi.org/10.1021/acsnano.7b07393. 8096–8101, https://doi.org/10.1039/C7NR00663B.
[121] C. Jang, J.H. Lee, A. Sahu, G. Tae, The synergistic effect of folate and RGD dual [146] D. Bitounis, H. Ali-Boucetta, B.H. Hong, D.-H. Min, K. Kostarelos, Prospects and
ligand of nanographene oxide on tumor targeting and photothermal therapy in challenges of graphene in biomedical applications, Adv. Mater. 25 (2013)
vivo, Nanoscale 7 (2015) 18584–18594, https://doi.org/10.1039/C5NR05067G. 2258–2268, https://doi.org/10.1002/adma.201203700.
[122] P. Wust, B. Hildebrandt, G. Sreenivasa, B. Rau, J. Gellermann, H. Riess, R. Felix, [147] K. Yang, L. Feng, X. Shi, Z. Liu, Nano-graphene in biomedicine: theranostic

23
J. Chen, et al. Biomaterials 237 (2020) 119827

applications, Chem. Soc. Rev. 42 (2013) 530–547, https://doi.org/10.1039/ D.-Y. Fan, H. Zhang, Black phosphorus-based field effect transistor devices for Ag
C2CS35342C. ions detection, Chin. Phys. B 27 (2018) 087308, , https://doi.org/10.1088/1674-
[148] X. Shi, H. Gong, Y. Li, C. Wang, L. Cheng, Z. Liu, Graphene-based magnetic 1056/27/8/087308.
plasmonic nanocomposite for dual bioimaging and photothermal therapy, [171] R. Wang, X. Jiang, S. Gao, J. Zhao, F. Zhang, W. Huang, T. Fan, W. Liang, Z. Li,
Biomaterials 34 (2013) 4786–4793, https://doi.org/10.1016/J.BIOMATERIALS. H. Huang, Z. Guo, H. Wang, Y. Zhang, X. Zhang, Z. Luo, H. Zhang, Unveiling the
2013.03.023. stimulated robust carrier lifetime of surface‐bound excitons and their photo-
[149] J. Ge, M. Lan, B. Zhou, W. Liu, L. Guo, H. Wang, Q. Jia, G. Niu, X. Huang, H. Zhou, response in InSe, Adv. Mater. Interfaces 6 (2019) 1900171, https://doi.org/10.
X. Meng, P. Wang, C.-S. Lee, W. Zhang, X. Han, A graphene quantum dot photo- 1002/admi.201900171.
dynamic therapy agent with high singlet oxygen generation, Nat. Commun. 5 [172] Z. Xie, C. Xing, W. Huang, T. Fan, Z. Li, J. Zhao, Y. Xiang, Z. Guo, J. Li, Z. Yang,
(2014) 4596, https://doi.org/10.1038/ncomms5596. B. Dong, J. Qu, D. Fan, H. Zhang, Ultrathin 2D nonlayered tellurium nanosheets:
[150] W.-S. Kuo, Y.-T. Shao, K.-S. Huang, T.-M. Chou, C.-H. Yang, Antimicrobial amino- facile liquid-phase exfoliation, characterization, and photoresponse with high
functionalized nitrogen-doped graphene quantum dots for eliminating multidrug- performance and enhanced stability, Adv. Funct. Mater. 28 (2018) 1705833,
resistant species in dual-modality photodynamic therapy and bioimaging under https://doi.org/10.1002/adfm.201705833.
two-photon excitation, ACS Appl. Mater. Interfaces 10 (2018) 14438–14446, [173] C. Xing, W. Huang, Z. Xie, J. Zhao, D. Ma, T. Fan, W. Liang, Y. Ge, B. Dong, J. Li,
https://doi.org/10.1021/acsami.8b01429. H. Zhang, Ultrasmall bismuth quantum dots: facile liquid-phase exfoliation,
[151] D. Shi, Y. Li, H. Dong, Y. Li, Graphene-based nanovehicles for photodynamic characterization, and application in high-performance UV–vis photodetector, ACS
medical therapy, Int. J. Nanomed. 10 (2015) 2451–2459, https://doi.org/10. Photonics 5 (2018) 621–629, https://doi.org/10.1021/acsphotonics.7b01211.
2147/IJN.S68600. [174] Z. Xie, F. Zhang, Z. Liang, T. Fan, Z. Li, X. Jiang, H. Chen, J. Li, H. Zhang,
[152] X. Wu, Y. Xing, K. Zeng, K. Huber, J.X. Zhao, Study of fluorescence quenching Revealing of the ultrafast third-order nonlinear optical response and enabled
ability of graphene oxide with a layer of rigid and tunable silica spacer, Langmuir photonic application in two-dimensional tin sulfide, Photon. Res. 7 (2019)
34 (2018) 603–611, https://doi.org/10.1021/acs.langmuir.7b03465. 494–502, https://doi.org/10.1364/PRJ.7.000494.
[153] Y. Ding, L. Zhou, X. Chen, Q. Wu, Z. Song, S. Wei, J. Zhou, J. Shen, Mutual sen- [175] M. Qiu, Z.T. Sun, D.K. Sang, X.G. Han, H. Zhang, C.M. Niu, Current progress in
sitization mechanism and self-degradation property of drug delivery system for in black phosphorus materials and their applications in electrochemical energy sto-
vitro photodynamic therapy, Int. J. Pharm. 498 (2016) 335–346, https://doi.org/ rage, Nanoscale 9 (2017) 13384–13403, https://doi.org/10.1039/C7NR03318D.
10.1016/J.IJPHARM.2015.12.044. [176] T. Xue, W. Liang, Y. Li, Y. Sun, Y. Xiang, Y. Zhang, Z. Dai, Y. Duo, L. Wu, K. Qi,
[154] Y. Cho, Y. Choi, Graphene oxide–photosensitizer conjugate as a redox-responsive B.N. Shivananju, L. Zhang, X. Cui, H. Zhang, Q. Bao, Ultrasensitive detection of
theranostic agent, Chem. Commun. 48 (2012) 9912–9914, https://doi.org/10. miRNA with an antimonene-based surface plasmon resonance sensor, Nat.
1039/c2cc35197h. Commun. 10 (2019) 28, https://doi.org/10.1038/s41467-018-07947-8.
[155] P. Rong, K. Yang, A. Srivastan, D.O. Kiesewetter, X. Yue, F. Wang, L. Nie, [177] J. Liu, X. Jiang, R. Zhang, Y. Zhang, L. Wu, W. Lu, J. Li, Y. Li, H. Zhang,
A. Bhirde, Z. Wang, Z. Liu, G. Niu, W. Wang, X. Chen, Photosensitizer loaded nano- MXene‐Enabled electrochemical microfluidic biosensor: applications toward
graphene for multimodality imaging guided tumor photodynamic therapy, multicomponent continuous monitoring in whole blood, Adv. Funct. Mater. 29
Theranostics 4 (2014) 229–239, https://doi.org/10.7150/thno.8070. (2019) 1807326, https://doi.org/10.1002/adfm.201807326.
[156] X. Yan, G. Niu, J. Lin, A.J. Jin, H. Hu, Y. Tang, Y. Zhang, A. Wu, J. Lu, S. Zhang, [178] L. Cheng, J. Liu, X. Gu, H. Gong, X. Shi, T. Liu, C. Wang, X. Wang, G. Liu, H. Xing,
P. Huang, B. Shen, X. Chen, Enhanced fluorescence imaging guided photodynamic W. Bu, B. Sun, Z. Liu, PEGylated WS 2 nanosheets as a multifunctional theranostic
therapy of sinoporphyrin sodium loaded graphene oxide, Biomaterials 42 (2015) agent for in vivo dual-modal CT/photoacoustic imaging guided photothermal
94–102, https://doi.org/10.1016/J.BIOMATERIALS.2014.11.040. therapy, Adv. Mater. 26 (2014) 1886–1893, https://doi.org/10.1002/adma.
[157] P. Kalluru, R. Vankayala, C.S. Chiang, K.C. Hwang, Nano-graphene oxide-medi- 201304497.
ated in vivo fluorescence imaging and bimodal photodynamic and photothermal [179] T. Liu, S. Shi, C. Liang, S. Shen, L. Cheng, C. Wang, X. Song, S. Goel, T.E. Barnhart,
destruction of tumors, Biomaterials 95 (2016) 1–10, https://doi.org/10.1016/j. W. Cai, Z. Liu, Iron oxide decorated MoS 2 nanosheets with double PEGylation for
biomaterials.2016.04.006. chelator-free radiolabeling and multimodal imaging guided photothermal therapy,
[158] D.-Y. Zhang, Y. Zheng, C.-P. Tan, J.-H. Sun, W. Zhang, L.-N. Ji, Z.-W. Mao, ACS Nano 9 (2015) 950–960, https://doi.org/10.1021/nn506757x.
Graphene oxide decorated with Ru(II)–Polyethylene glycol complex for lysosome- [180] L.V. Wang, S. Hu, Photoacoustic tomography: in vivo imaging from organelles to
targeted imaging and photodynamic/photothermal therapy, ACS Appl. Mater. organs, Science 335 (2012) 1458–1462, https://doi.org/10.1126/science.
Interfaces 9 (2017) 6761–6771, https://doi.org/10.1021/acsami.6b13808. 1216210.
[159] F. Zhao, H. Meng, L. Yan, B. Wang, Y. Zhao, Nanosurface chemistry and dose [181] S. Wang, X. Li, Y. Chen, X. Cai, H. Yao, W. Gao, Y. Zheng, X. An, J. Shi, H. Chen, A
govern the bioaccumulation and toxicity of carbon nanotubes, metal nanomater- facile one-pot synthesis of a two-dimensional MoS 2/Bi 2 S 3 composite theranostic
ials and quantum dots in vivo, Sci. Bull. 60 (2015) 3–20, https://doi.org/10.1007/ nanosystem for multi-modality tumor imaging and therapy, Adv. Mater. 27 (2015)
s11434-014-0700-0. 2775–2782, https://doi.org/10.1002/adma.201500870.
[160] S. Kang, M. Herzberg, D.F. Rodrigues, M. Elimelech, Antibacterial effects of carbon [182] T. Liu, C. Wang, X. Gu, H. Gong, L. Cheng, X. Shi, L. Feng, B. Sun, Z. Liu, Drug
nanotubes: size does matter!, Langmuir 24 (2008) 6409–6413, https://doi.org/10. delivery with PEGylated MoS 2 nano-sheets for combined photothermal and che-
1021/la800951v. motherapy of cancer, Adv. Mater. 26 (2014) 3433–3440, https://doi.org/10.
[161] M. Li, Z. Luo, Y. Zhao, Recent advancements in 2D nanomaterials for cancer 1002/adma.201305256.
therapy, Sci. China Chem. 61 (2018) 1214–1226, https://doi.org/10.1007/ [183] Y. Ma, H. Wang, D. Yan, Y. Wei, Y. Cao, P. Yi, H. Zhang, Z. Deng, J. Dai, X. Liu,
s11426-018-9294-9. J. Luo, Z. Zhang, S. Sun, H. Guo, Magnetic resonance imaging revealed splenic
[162] W. Tao, X. Ji, X. Xu, M.A. Islam, Z. Li, S. Chen, P.E. Saw, H. Zhang, Z. Bharwani, targeting of canine parvovirus capsid protein VP2, Sci. Rep. 6 (2016) 23392,
Z. Guo, J. Shi, O.C. Farokhzad, Antimonene quantum dots: synthesis and appli- https://doi.org/10.1038/srep23392.
cation as near-infrared photothermal agents for effective cancer therapy, Angew. [184] N. Shadjou, M. Hasanzadeh, Graphene and its nanostructure derivatives for use in
Chem. Int. Ed. 56 (2017) 11896–11900, https://doi.org/10.1002/anie. bone tissue engineering: recent advances, J. Biomed. Mater. Res. 104 (2016)
201703657. 1250–1275, https://doi.org/10.1002/jbm.a.35645.
[163] C. Xing, Z. Xie, Z. Liang, W. Liang, T. Fan, J.S. Ponraj, S.C. Dhanabalan, D. Fan, [185] Z. Xie, S. Chen, Y. Duo, Y. Zhu, T. Fan, Q. Zou, M. Qu, Z. Lin, J. Zhao, Y. Li, L. Liu,
H. Zhang, 2D nonlayered selenium nanosheets: facile synthesis, photo- S. Bao, H. Chen, D. Fan, H. Zhang, Biocompatible two-dimensional titanium na-
luminescence, and ultrafast photonics, Adv. Opt. Mater. 5 (2017) 1700884, , nosheets for multimodal imaging-guided cancer theranostics, ACS Appl. Mater.
https://doi.org/10.1002/adom.201700884. Interfaces 11 (2019) 22129–22140, https://doi.org/10.1021/acsami.9b04628.
[164] C. Murugan, V. Sharma, R.K. Murugan, G. Malaimegu, A. Sundaramurthy, Two- [186] J. Liu, W. Bu, J. Shi, Chemical design and synthesis of functionalized probes for
dimensional cancer theranostic nanomaterials: synthesis, surface functionalization imaging and treating tumor hypoxia, Chem. Rev. 117 (2017) 6160–6224, https://
and applications in photothermal therapy, J. Contr. Release 299 (2019) 1–20, doi.org/10.1021/acs.chemrev.6b00525.
https://doi.org/10.1016/J.JCONREL.2019.02.015. [187] F. Jiang, A.M. Robin, M. Katakowski, L. Tong, M. Espiritu, G. Singh, M. Chopp,
[165] M. Qiu, W.X. Ren, T. Jeong, M. Won, G.Y. Park, D.K. Sang, L.-P. Liu, H. Zhang, Photodynamic therapy with photofrin in combination with Buthionine
J.S. Kim, Omnipotent phosphorene: a next-generation, two-dimensional nano- Sulfoximine (BSO) of human glioma in the nude rat, Laser Med. Sci. 18 (2003)
platform for multidisciplinary biomedical applications, Chem. Soc. Rev. 47 (2018) 128–133, https://doi.org/10.1007/s10103-003-0269-3.
5588–5601, https://doi.org/10.1039/C8CS00342D. [188] T. Kiesslich, K. Plaetzer, C.B. Oberdanner, J. Berlanda, F.J. Obermair, B. Krammer,
[166] T. Fan, Y. Zhou, M. Qiu, H. Zhang, Black phosphorus: a novel nanoplatform with Differential effects of glucose deprivation on the cellular sensitivity towards
potential in the field of bio-photonic nanomedicine, J. Innov. Opt. Health Sci. 11 photodynamic treatment-based production of reactive oxygen species and apop-
(2018) 1830003, , https://doi.org/10.1142/S1793545818300033. tosis-induction, FEBS Lett. 579 (2005) 185–190, https://doi.org/10.1016/j.
[167] M. Luo, T. Fan, Y. Zhou, H. Zhang, L. Mei, 2D black phosphorus–based biomedical febslet.2004.11.073.
applications, Adv. Funct. Mater. 29 (2019) 1808306, , https://doi.org/10.1002/ [189] H. Fan, G. Yan, Z. Zhao, X. Hu, W. Zhang, H. Liu, X. Fu, T. Fu, X.-B. Zhang, W. Tan,
adfm.201808306. A smart photosensitizer-manganese dioxide nanosystem for enhanced photo-
[168] T. Fan, Z. Xie, W. Huang, Z. Li, H. Zhang, Two-dimensional non-layered selenium dynamic therapy by reducing glutathione levels in cancer cells, Angew. Chem. Int.
nanoflakes: facile fabrications and applications for self-powered photo-detector, Ed. 55 (2016) 5477–5482, https://doi.org/10.1002/anie.201510748.
Nanotechnology 30 (2019) 114002, , https://doi.org/10.1088/1361-6528/aafc0f. [190] Y. Chen, D. Ye, M. Wu, H. Chen, L. Zhang, J. Shi, L. Wang, Break-up of two-
[169] W. Huang, Z. Xie, T. Fan, J. Li, Y. Wang, L. Wu, D. Ma, Z. Li, Y. Ge, Z.N. Huang, dimensional MnO2 nanosheets promotes ultrasensitive pH-triggered theranostics
X. Dai, Y. Xiang, J. Li, X. Zhu, H. Zhang, Black-phosphorus-analogue tin mono- of cancer, Adv. Mater. 26 (2014) 7019–7026, https://doi.org/10.1002/adma.
sulfide: an emerging optoelectronic two-dimensional material for high-perfor- 201402572.
mance photodetection with improved stability under ambient/harsh conditions, J. [191] Y. Chen, H. Chen, S. Zhang, F. Chen, S. Sun, Q. He, M. Ma, X. Wang, H. Wu,
Mater. Chem. C. 6 (2018) 9582–9593, https://doi.org/10.1039/C8TC03284J. L. Zhang, L. Zhang, J. Shi, Structure-property relationships in manganese oxide -
[170] H.-D. Wang, D.K. Sang, Z.-N. Guo, R. Cao, J.-L. Zhao, M.N. Ullah Shah, T.-J. Fan, mesoporous silica nanoparticles used for T 1-weighted MRI and simultaneous anti-

24
J. Chen, et al. Biomaterials 237 (2020) 119827

cancer drug delivery, Biomaterials 33 (2012) 2388–2398, https://doi.org/10. [215] J. Shi, Z. Li, D.K. Sang, Y. Xiang, J. Li, S. Zhang, H. Zhang, THz photonics in two
1016/j.biomaterials.2011.11.086. dimensional materials and metamaterials: properties, devices and prospects, J.
[192] W. Fan, W. Bu, B. Shen, Q. He, Z. Cui, Y. Liu, X. Zheng, K. Zhao, J. Shi, Intelligent Mater. Chem. C. 6 (2018) 1291–1306, https://doi.org/10.1039/C7TC05460B.
MnO 2 nanosheets anchored with upconversion nanoprobes for concurrent pH-/H [216] Z. Guo, S. Chen, Z. Wang, Z. Yang, F. Liu, Y. Xu, J. Wang, Y. Yi, H. Zhang, L. Liao,
2 O 2 -responsive UCL imaging and oxygen-elevated synergetic therapy, Adv. P.K. Chu, X.-F. Yu, Metal-ion-modified black phosphorus with enhanced stability
Mater. 27 (2015) 4155–4161, https://doi.org/10.1002/adma.201405141. and transistor performance, Adv. Mater. 29 (2017) 1703811, https://doi.org/10.
[193] C. Zhang, W.-H. Chen, L.-H. Liu, W.-X. Qiu, W.-Y. Yu, X.-Z. Zhang, An O 2 self- 1002/adma.201703811.
supplementing and reactive-oxygen-species-circulating amplified nanoplatform [217] Y. Ge, S. Chen, Y. Xu, Z. He, Z. Liang, Y. Chen, Y. Song, D. Fan, K. Zhang, H. Zhang,
via H 2 O/H 2 O 2 splitting for tumor imaging and photodynamic therapy, Adv. Few-layer selenium-doped black phosphorus: synthesis, nonlinear optical prop-
Funct. Mater. 27 (2017) 1700626, , https://doi.org/10.1002/adfm.201700626. erties and ultrafast photonics applications, J. Mater. Chem. C. 5 (2017)
[194] D. Zeng, L. Wang, L. Tian, S. Zhao, X. Zhang, H. Li, Synergistic photothermal/ 6129–6135, https://doi.org/10.1039/C7TC01267E.
photodynamic suppression of prostatic carcinoma by targeted biodegradable MnO [218] J. Du, M. Zhang, Z. Guo, J. Chen, X. Zhu, G. Hu, P. Peng, Z. Zheng, H. Zhang,
2 nanosheets, Drug Deliv. 26 (2019) 661–672, https://doi.org/10.1080/ Phosphorene quantum dot saturable absorbers for ultrafast fiber lasers, Sci. Rep. 7
10717544.2019.1631409. (2017) 42357, https://doi.org/10.1038/srep42357.
[195] H. Chen, T. Liu, Z. Su, L. Shang, G. Wei, 2D transition metal dichalcogenide na- [219] S.C. Dhanabalan, J.S. Ponraj, Z. Guo, S. Li, Q. Bao, H. Zhang, Emerging trends in
nosheets for photo/thermo-based tumor imaging and therapy, Nanoscale Horizons phosphorene fabrication towards next generation devices, Adv. Sci. 4 (2017)
3 (2018) 74–89, https://doi.org/10.1039/C7NH00158D. 1600305, , https://doi.org/10.1002/advs.201600305.
[196] Z. Xie, D. Wang, T. Fan, C. Xing, Z. Li, W. Tao, L. Liu, S. Bao, D. Fan, H. Zhang, [220] X. Chen, G. Xu, X. Ren, Z. Li, X. Qi, K. Huang, H. Zhang, Z. Huang, J. Zhong, A
Black phosphorus analogue tin sulfide nanosheets: synthesis and application as black/red phosphorus hybrid as an electrode material for high-performance Li-ion
near-infrared photothermal agents and drug delivery platforms for cancer therapy, batteries and supercapacitors, J. Mater. Chem. A. 5 (2017) 6581–6588, https://
J. Mater. Chem. B. 6 (2018) 4747–4755, https://doi.org/10.1039/C8TB00729B. doi.org/10.1039/C7TA00455A.
[197] H. Li, J. Wu, Z. Yin, H. Zhang, Preparation and applications of mechanically ex- [221] Y. Xu, W. Wang, Y. Ge, H. Guo, X. Zhang, S. Chen, Y. Deng, Z. Lu, H. Zhang,
foliated single-layer and multilayer MoS 2 and WSe 2 nanosheets, Acc. Chem. Res. Stabilization of black phosphorous quantum dots in PMMA nanofiber film and
47 (2014) 1067–1075, https://doi.org/10.1021/ar4002312. broadband nonlinear optics and ultrafast photonics application, Adv. Funct.
[198] Z. Yin, H. Li, H. Li, L. Jiang, Y. Shi, Y. Sun, G. Lu, Q. Zhang, X. Chen, H. Zhang, Mater. 27 (2017) 1702437, https://doi.org/10.1002/adfm.201702437.
Single-layer MoS 2 phototransistors, ACS Nano 6 (2012) 74–80, https://doi.org/ [222] H. Liu, A.T. Neal, Z. Zhu, Z. Luo, X. Xu, D. Tománek, P.D. Ye, Phosphorene: an
10.1021/nn2024557. unexplored 2D semiconductor with a high hole mobility, ACS Nano 8 (2014)
[199] Q. He, Z. Zeng, Z. Yin, H. Li, S. Wu, X. Huang, H. Zhang, Fabrication of flexible 4033–4041, https://doi.org/10.1021/nn501226z.
MoS 2 thin-film transistor arrays for practical gas-sensing applications, Small 8 [223] X. Ling, H. Wang, S. Huang, F. Xia, M.S. Dresselhaus, The renaissance of black
(2012) 2994–2999, https://doi.org/10.1002/smll.201201224. phosphorus, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) 4523–4530, https://doi.
[200] T. Liu, Z. Liu, 2D MoS 2 nanostructures for biomedical applications, Adv. Health org/10.1073/pnas.1416581112.
Mater. 7 (2018) 1701158, https://doi.org/10.1002/adhm.201701158. [224] M. Qiu, D. Wang, W. Liang, L. Liu, Y. Zhang, X. Chen, D.K. Sang, C. Xing, Z. Li,
[201] T. Liu, C. Wang, W. Cui, H. Gong, C. Liang, X. Shi, Z. Li, B. Sun, Z. Liu, Combined B. Dong, F. Xing, D. Fan, S. Bao, H. Zhang, Y. Cao, Novel concept of the smart NIR-
photothermal and photodynamic therapy delivered by PEGylated MoS 2 na- light-controlled drug release of black phosphorus nanostructure for cancer
nosheets, Nanoscale 6 (2014) 11219–11225, https://doi.org/10.1039/ therapy, Proc. Natl. Acad. Sci. U. S. A. 115 (2018) 501–506, https://doi.org/10.
C4NR03753G. 1073/pnas.1714421115.
[202] D.-K. Ji, Y. Zhang, Y. Zang, J. Li, G.-R. Chen, X.-P. He, H. Tian, Targeted in- [225] Z. Sun, H. Xie, S. Tang, X.-F. Yu, Z. Guo, J. Shao, H. Zhang, H. Huang, H. Wang,
tracellular production of reactive oxygen species by a 2D molybdenum disulfide P.K. Chu, Ultrasmall black phosphorus quantum dots: synthesis and use as pho-
glycosheet, Adv. Mater. 28 (2016) 9356–9363, https://doi.org/10.1002/adma. tothermal agents, Angew. Chem. Int. Ed. 54 (2015) 11526–11530, https://doi.
201602748. org/10.1002/anie.201506154.
[203] S. Kapri, S. Bhattacharyya, Molybdenum sulfide–reduced graphene oxide p–n [226] C. Xing, S. Chen, M. Qiu, X. Liang, Q. Liu, Q. Zou, Z. Li, Z. Xie, D. Wang, B. Dong,
heterojunction nanosheets with anchored oxygen generating manganese dioxide L. Liu, D. Fan, H. Zhang, Conceptually novel black phosphorus/cellulose
nanoparticles for enhanced photodynamic therapy, Chem. Sci. 9 (2018) Hydrogels as promising photothermal agents for effective cancer therapy, Adv.
8982–8989, https://doi.org/10.1039/C8SC02508H. Health Mater. 7 (2018) 1701510, , https://doi.org/10.1002/adhm.201701510.
[204] F. Meng, J. Li, S.K. Cushing, M. Zhi, N. Wu, Solar Hydrogen generation by na- [227] Z. Sun, Y. Zhao, Z. Li, H. Cui, Y. Zhou, W. Li, W. Tao, H. Zhang, H. Wang, P.K. Chu,
noscale p–n junction of p -type molybdenum disulfide/n -type nitrogen-doped X.-F. Yu, TiL 4 -coordinated black phosphorus quantum dots as an efficient contrast
reduced graphene oxide, J. Am. Chem. Soc. 135 (2013) 10286–10289, https://doi. agent for in vivo photoacoustic imaging of cancer, Small 13 (2017) 1602896,
org/10.1021/ja404851s. https://doi.org/10.1002/smll.201602896.
[205] L. Yang, J. Wang, S. Yang, Q. Lu, P. Li, N. Li, Rod-shape MSN@MoS2 nanoplatform [228] X. Qian, Z. Gu, Y. Chen, Two-dimensional black phosphorus nanosheets for ther-
for FL/MSOT/CT imaging-guided photothermal and photodynamic therapy, anostic nanomedicine, Mater. Horizons. 4 (2017) 800–816, https://doi.org/10.
Theranostics 9 (2019) 3992–4005, https://doi.org/10.7150/thno.32715. 1039/C7MH00305F.
[206] J. Wang, G. Zhu, M. You, E. Song, M.I. Shukoor, K. Zhang, M.B. Altman, Y. Chen, [229] W. Tao, X. Zhu, X. Yu, X. Zeng, Q. Xiao, X. Zhang, X. Ji, X. Wang, J. Shi, H. Zhang,
Z. Zhu, C.Z. Huang, W. Tan, Assembly of aptamer switch probes and photo- L. Mei, Black phosphorus nanosheets as a robust delivery platform for cancer
sensitizer on gold nanorods for targeted photothermal and photodynamic cancer theranostics, Adv. Mater. 29 (2017) 1603276, https://doi.org/10.1002/adma.
therapy, ACS Nano 6 (2012) 5070–5077, https://doi.org/10.1021/nn300694v. 201603276.
[207] Y. Yong, L. Zhou, Z. Gu, L. Yan, G. Tian, X. Zheng, X. Liu, X. Zhang, J. Shi, [230] F. Yin, K. Hu, S. Chen, D. Wang, J. Zhang, M. Xie, D. Yang, M. Qiu, H. Zhang, Z. Li,
W. Cong, W. Yin, Y. Zhao, WS 2 nanosheet as a new photosensitizer carrier for Black phosphorus quantum dot based novel siRNA delivery systems in human
combined photodynamic and photothermal therapy of cancer cells, Nanoscale 6 pluripotent teratoma PA-1 cells, J. Mater. Chem. B. 5 (2017) 5433–5440, https://
(2014) 10394–10403, https://doi.org/10.1039/C4NR02453B. doi.org/10.1039/C7TB01068K.
[208] L.-S. Lin, Z.-X. Cong, J. Li, K.-M. Ke, S.-S. Guo, H.-H. Yang, G.-N. Chen, Graphitic- [231] J.R. Choi, K.W. Yong, J.Y. Choi, A. Nilghaz, Y. Lin, J. Xu, X. Lu, Black phosphorus
phase C3N4 nanosheets as efficient photosensitizers and pH-responsive drug na- and its biomedical applications, Theranostics 8 (2018) 1005–1026, https://doi.
nocarriers for cancer imaging and therapy, J. Mater. Chem. B. 2 (2014) org/10.7150/thno.22573.
1031–1037, https://doi.org/10.1039/c3tb21479f. [232] D.L. Childers, J. Corman, M. Edwards, J.J. Elser, Sustainability challenges of
[209] Z. Ma, M. Zhang, X. Jia, J. Bai, Y. Ruan, C. Wang, X. Sun, X. Jiang, Fe III -doped phosphorus and food: solutions from closing the human phosphorus cycle,
two-dimensional C 3 N 4 nanofusiform: a new O 2 -evolving and mitochondria- Bioscience 61 (2011) 117–124, https://doi.org/10.1525/bio.2011.61.2.6.
targeting photodynamic agent for MRI and enhanced antitumor therapy, Small 12 [233] J. Shao, H. Xie, H. Huang, Z. Li, Z. Sun, Y. Xu, Q. Xiao, X.-F. Yu, Y. Zhao, H. Zhang,
(2016) 5477–5487, https://doi.org/10.1002/smll.201601681. H. Wang, P.K. Chu, Biodegradable black phosphorus-based nanospheres for in vivo
[210] D.-W. Zheng, B. Li, C.-X. Li, J.-X. Fan, Q. Lei, C. Li, Z. Xu, X.-Z. Zhang, Carbon-dot- photothermal cancer therapy, Nat. Commun. 7 (2016) 12967, https://doi.org/10.
decorated carbon nitride nanoparticles for enhanced photodynamic therapy 1038/ncomms12967.
against hypoxic tumor via water splitting, ACS Nano 10 (2016) 8715–8722, [234] H. Wang, X. Yang, W. Shao, S. Chen, J. Xie, X. Zhang, J. Wang, Y. Xie, Ultrathin
https://doi.org/10.1021/acsnano.6b04156. black phosphorus nanosheets for efficient singlet oxygen generation, J. Am. Chem.
[211] J. Dai, J. Song, Y. Qiu, J. Wei, Z. Hong, L. Li, H. Yang, Gold nanoparticle-decorated Soc. 137 (2015) 11376–11382, https://doi.org/10.1021/jacs.5b06025.
g-C 3 N 4 nanosheets for controlled generation of reactive oxygen species upon 670 [235] R. Lv, D. Yang, P. Yang, J. Xu, F. He, S. Gai, C. Li, Y. Dai, G. Yang, J. Lin,
nm laser illumination, ACS Appl. Mater. Interfaces 11 (2019) 10589–10596, Integration of upconversion nanoparticles and ultrathin black phosphorus for ef-
https://doi.org/10.1021/acsami.9b01307. ficient photodynamic theranostics under 808 nm near-infrared light irradiation,
[212] Z. Chu, J. Liu, Z. Guo, H. Zhang, 2 μm passively Q-switched laser based on black Chem. Mater. 28 (2016) 4724–4734, https://doi.org/10.1021/acs.chemmater.
phosphorus, Opt. Mater. Express 6 (2016) 2374–2379, https://doi.org/10.1364/ 6b01720.
OME.6.002374. [236] D. Yang, G. Yang, P. Yang, R. Lv, S. Gai, C. Li, F. He, J. Lin, Assembly of Au
[213] Y. Xu, J. Yuan, K. Zhang, Y. Hou, Q. Sun, Y. Yao, S. Li, Q. Bao, H. Zhang, Y. Zhang, plasmonic photothermal agent and iron oxide nanoparticles on ultrathin black
Field-induced n-doping of black phosphorus for CMOS compatible 2D logic elec- phosphorus for targeted photothermal and photodynamic cancer therapy, Adv.
tronics with high electron mobility, Adv. Funct. Mater. 27 (2017) 1702211, Funct. Mater. 27 (2017) 1700371, https://doi.org/10.1002/adfm.201700371.
https://doi.org/10.1002/adfm.201702211. [237] X. Yang, D. Wang, J. Zhu, L. Xue, C. Ou, W. Wang, M. Lu, X. Song, X. Dong,
[214] Y. Xu, X.-F. Jiang, Y. Ge, Z. Guo, Z. Zeng, Q.-H. Xu, H. Zhang, X.-F. Yu, D. Fan, Functional black phosphorus nanosheets for mitochondria-targeting photo-
Size-dependent nonlinear optical properties of black phosphorus nanosheets and thermal/photodynamic synergistic cancer therapy, Chem. Sci. 10 (2019)
their applications in ultrafast photonics, J. Mater. Chem. C. 5 (2017) 3007–3013, 3779–3785, https://doi.org/10.1039/C8SC04844D.
https://doi.org/10.1039/C7TC00071E. [238] J. Liu, P. Du, T. Liu, B.J. Córdova Wong, W. Wang, H. Ju, J. Lei, A black

25
J. Chen, et al. Biomaterials 237 (2020) 119827

phosphorus/manganese dioxide nanoplatform: oxygen self-supply monitoring, P.A. Bunn, S.F. Kolhouse, New anti‐lung‐cancer antibody cluster 12 reacts with
photodynamic therapy enhancement and feedback, Biomaterials 192 (2019) human folate receptors present on adenocarcinoma, Int. J. Canc. 57 (1994) 89–95,
179–188, https://doi.org/10.1016/J.BIOMATERIALS.2018.10.018. https://doi.org/10.1002/ijc.2910570719.
[239] Y. Chen, Y. Wu, B. Sun, S. Liu, H. Liu, Two-dimensional nanomaterials for cancer [262] G. Toffoli, C. Cernigoi, A. Russo, A. Gallo, M. Bagnoli, M. Boiocchi, Overexpression
nanotheranostics, Small 13 (2017) 1603446, , https://doi.org/10.1002/smll. of folate binding protein in ovarian cancers, Int. J. Canc. 74 (1997) 193–198,
201603446. https://doi.org/10.1002/(SICI)1097-0215(19970422)74:2<193::AID-IJC10>3.
[240] T. Hu, X. Mei, Y. Wang, X. Weng, R. Liang, M. Wei, Two-dimensional nanoma- 0.CO;2-F.
terials: fascinating materials in biomedical field, Sci. Bull. 64 (2019) 1707–1727, [263] M.R. Hamblin, E.L. Newman, Photosensitizer targeting in photodynamic therapy
https://doi.org/10.1016/j.scib.2019.09.021. II. Conjugates of haematoporphyrin with serum lipoproteins, J. Photochem.
[241] W. Liu, Y.-M. Wang, Y.-H. Li, S.-J. Cai, X.-B. Yin, X.-W. He, Y.-K. Zhang, Photobiol. B Biol. 26 (1994) 147–157, https://doi.org/10.1016/1011-1344(94)
Fluorescent imaging-guided chemotherapy-and-photodynamic dual therapy with 07036-9.
nanoscale porphyrin metal-organic framework, Small 13 (2017) 1603459, https:// [264] U. Schmidt-Erfurth, H. Diddens, R. Birngruber, T. Hasan, Photodynamic targeting
doi.org/10.1002/smll.201603459. of human retinoblastoma cells using covalent low-density lipoprotein conjugates,
[242] J. Park, Q. Jiang, D. Feng, L. Mao, H.C. Zhou, Size-controlled synthesis of por- Br. J. Canc. 75 (1997) 54–61, https://doi.org/10.1038/bjc.1997.9.
phyrinic metal-organic framework and functionalization for targeted photo- [265] S.V. Lutsenko, N.B. Feldman, G.V. Finakova, G.A. Posypanova, S.E. Severin,
dynamic therapy, J. Am. Chem. Soc. 138 (2016) 3518–3525, https://doi.org/10. K.G. Skryabin, M.P. Kirpichnikov, E.A. Lukyanets, G.N. Vorozhtsov, Targeting
1021/jacs.6b00007. phthalocyanines to tumor cells using epidermal growth factor conjugates, Tumor
[243] K. Lu, T. Aung, N. Guo, R. Weichselbaum, W. Lin, Nanoscale metal-organic fra- Biol. 20 (1999) 218–224, https://doi.org/10.1159/000030066.
meworks for therapeutic, imaging, and sensing applications, Adv. Mater. 30 [266] M.D. Savellano, T. Hasan, Photochemical targeting of epidermal growth factor
(2018) 1707634, , https://doi.org/10.1002/adma.201707634. receptor: a mechanistic study, Clin. Canc. Res. 11 (2005) 1658–1668, https://doi.
[244] K. Lu, C. He, W. Lin, Nanoscale metal-organic framework for highly effective org/10.1158/1078-0432.CCR-04-1902.
photodynamic therapy of resistant head and neck cancer, J. Am. Chem. Soc. 136 [267] M. Sibrian-Vazquez, T.J. Jensen, M.G.H. Vicente, Porphyrin−Retinamides:
(2014) 16712–16715, https://doi.org/10.1021/ja508679h. synthesis and cellular studies, Bioconjugate Chem. 18 (2007) 1185–1193, https://
[245] K. Lu, C. He, W. Lin, A chlorin-based nanoscale metal-organic framework for doi.org/10.1021/bc0700382.
photodynamic therapy of colon cancers, J. Am. Chem. Soc. 137 (2015) [268] M.D. Savellano, B.W. Pogue, P.J. Hoopes, E.S. Vitetta, K.D. Paulsen, Multiepitope
7600–7603, https://doi.org/10.1021/jacs.5b04069. HER2 targeting enhances photoimmunotherapy of HER2-overexpressing cancer
[246] G. Lan, K. Ni, Z. Xu, S.S. Veroneau, Y. Song, W. Lin, Nanoscale metal–organic cells with pyropheophorbide-a immunoconjugates, Canc. Res. 65 (2005)
framework overcomes hypoxia for photodynamic therapy primed cancer im- 6371–6379, https://doi.org/10.1158/0008-5472.CAN-05-0426.
munotherapy, J. Am. Chem. Soc. 140 (2018) 5670–5673, https://doi.org/10. [269] G.C. Bolfarini, M.P. Siqueira-Moura, G.J.F. Demets, P.C. Morais, A.C. Tedesco, In
1021/jacs.8b01072. vitro evaluation of combined hyperthermia and photodynamic effects using
[247] M. He, Y. Chen, C. Tao, Q. Tian, L. An, J. Lin, Q. Tian, H. Yang, S. Yang, magnetoliposomes loaded with cucurbituril zinc phthalocyanine complex on
Mn–Porphyrin-Based metal–organic framework with high longitudinal relaxivity melanoma, J. Photochem. Photobiol. B Biol. 115 (2012) 1–4, https://doi.org/10.
for magnetic resonance imaging guidance and oxygen self-supplementing photo- 1016/j.jphotobiol.2012.05.009.
dynamic therapy, ACS Appl. Mater. Interfaces 11 (2019) 41946–41956, https:// [270] V. Sol, V. Chaleix, Y. Champavier, R. Granet, Y.M. Huang, P. Krausz, Glycosyl bis-
doi.org/10.1021/acsami.9b15083. porphyrin conjugates: synthesis and potential application in PDT, Bioorg. Med.
[248] X. Li, B. De Zheng, X.H. Peng, S.Z. Li, J.W. Ying, Y. Zhao, J.D. Huang, J. Yoon, Chem. 14 (2006) 7745–7760, https://doi.org/10.1016/j.bmc.2006.08.004.
Phthalocyanines as medicinal photosensitizers: developments in the last five years, [271] J.V. Frangioni, In vivo near-infrared fluorescence imaging, Curr. Opin. Chem. Biol.
Coord. Chem. Rev. 379 (2019) 147–160, https://doi.org/10.1016/j.ccr.2017.08. 7 (2003) 626–634, https://doi.org/10.1016/J.CBPA.2003.08.007.
003. [272] Z. Zhou, J. Song, L. Nie, X. Chen, Reactive oxygen species generating systems
[249] C.S. Jin, L. Cui, F. Wang, J. Chen, G. Zheng, Targeting-triggered porphysome meeting challenges of photodynamic cancer therapy, Chem. Soc. Rev. 45 (2016)
nanostructure disruption for activatable photodynamic therapy, Adv. Health 6597–6626, https://doi.org/10.1039/C6CS00271D.
Mater. 3 (2014) 1240–1249, https://doi.org/10.1002/adhm.201300651. [273] C. Wang, L. Cheng, Z. Liu, Upconversion nanoparticles for photodynamic therapy
[250] X. Li, C.Y. Kim, S. Lee, D. Lee, H.M. Chung, G. Kim, S.H. Heo, C. Kim, K.S. Hong, and other cancer therapeutics, Theranostics 3 (2013) 317–330, https://doi.org/
J. Yoon, Nanostructured phthalocyanine assemblies with protein-driven switch- 10.7150/thno.5284.
able photoactivities for biophotonic imaging and therapy, J. Am. Chem. Soc. 139 [274] B.W. Pogue, T. Hasan, A theoretical study of light fractionation and dose-rate
(2017) 10880–10886, https://doi.org/10.1021/jacs.7b05916. effects in photodynamic therapy, Radiat. Res. 147 (1997) 551–559, https://doi.
[251] X. Li, S. Yu, D. Lee, G. Kim, B. Lee, Y. Cho, B.Y. Zheng, M.R. Ke, J.D. Huang, org/10.2307/3579621.
K.T. Nam, X. Chen, J. Yoon, Facile supramolecular approach to nucleic-acid-driven [275] M. Ascencio, J.P. Estevez, M. Delemer, M.O. Farine, P. Collinet, S. Mordon,
activatable nanotheranostics that overcome drawbacks of photodynamic therapy, Comparison of continuous and fractionated illumination during hexaminolaevu-
ACS Nano 12 (2018) 681–688, https://doi.org/10.1021/acsnano.7b07809. linate-photodynamic therapy, Photodiagnosis Photodyn. Ther. 5 (2008) 210–216,
[252] X. Li, D. Lee, J.-D. Huang, J. Yoon, Phthalocyanine-assembled nanodots as pho- https://doi.org/10.1016/J.PDPDT.2008.09.004.
tosensitizers for highly efficient Type I photoreactions in photodynamic therapy, [276] B.W. Henderson, T.M. Busch, L.A. Vaughan, N.P. Frawley, D. Babich, T.A. Sosa,
Angew. Chem. Int. Ed. 57 (2018) 9885–9890, https://doi.org/10.1002/anie. J.D. Zollo, A.S. Dee, M.T. Cooper, D.A. Bellnier, W.R. Greco, A.R. Oseroff,
201806551. Photofrin photodynamic therapy can significantly deplete or preserve oxygenation
[253] X. Li, S. Yu, Y. Lee, T. Guo, N. Kwon, D. Lee, S.C. Yeom, Y. Cho, G. Kim, J.- in human basal cell carcinomas during treatment, depending on fluence rate,
D. Huang, S. Choi, K.T. Nam, J. Yoon, In vivo albumin traps photosensitizer Canc. Res. 60 (2000) 525–529 http://www.ncbi.nlm.nih.gov/pubmed/10676629.
monomers from self-assembled phthalocyanine nanovesicles: a facile and switch- [277] J.C. Finlay, D.L. Conover, E.L. Hull, T.H. Foster, Porphyrin bleaching and PDT-
able theranostic approach, J. Am. Chem. Soc. 141 (2019) 1366–1372, https://doi. induced spectral changes are irradiance dependent in ALA-sensitized normal rat
org/10.1021/jacs.8b12167. skin in vivo, Photochem. Photobiol. 73 (2007) 54–63, https://doi.org/10.1562/
[254] D. Li, X.-Z. Wang, L.-F. Yang, S.-C. Li, Q.-Y. Hu, X. Li, B.-Y. Zheng, M.-R. Ke, J.- 0031-8655(2001)0730054PBAPIS2.0.CO2.
D. Huang, Size-tunable targeting-triggered nanophotosensitizers based on self- [278] T.A. Middelburg, F. Van Zaane, H.S. De Bruijn, A. Van Der Ploeg-van den Heuvel,
assembly of a phthalocyanine–biotin conjugate for photodynamic therapy, ACS H.J.C.M. Sterenborg, H.A.M. Neumann, E.R.M. De Haas, D.J. Robinson,
Appl. Mater. Interfaces 11 (2019) 36435–36443, https://doi.org/10.1021/acsami. Fractionated illumination at low fluence rate photodynamic therapy in mice,
9b13861. Photochem. Photobiol. 86 (2010) 1140–1146, https://doi.org/10.1111/j.1751-
[255] J.F. Lovell, C.S. Jin, E. Huynh, T.D. MacDonald, W. Cao, G. Zheng, Enzymatic 1097.2010.00760.x.
regioselection for the synthesis and biodegradation of porphysome nanovesicles, [279] Q. Chen, Z. Huang, H. Chen, H. Shapiro, J. Beckers, F.W. Hetzel, Improvement of
Angew. Chem. Int. Ed. 51 (2012) 2429–2433, https://doi.org/10.1002/anie. tumor response by manipulation of tumor oxygenation during photodynamic
201108280. therapy, Photochem. Photobiol. 76 (2007) 197–203, https://doi.org/10.1562/
[256] K. Huang, Z. Li, J. Lin, G. Han, P. Huang, Two-dimensional transition metal car- 0031-8655(2002)0760197IOTRBM2.0.CO2.
bides and nitrides (MXenes) for biomedical applications, Chem. Soc. Rev. 47 [280] X. Li, N. Kwon, T. Guo, Z. Liu, J. Yoon, Innovative strategies for hypoxic-tumor
(2018) 5109–5124, https://doi.org/10.1039/c7cs00838d. photodynamic therapy, Angew. Chem. Int. Ed. 57 (2018) 11522–11531, https://
[257] G. Liu, J. Zou, Q. Tang, X. Yang, Y. Zhang, Q. Zhang, W. Huang, P. Chen, J. Shao, doi.org/10.1002/anie.201805138.
X. Dong, Surface modified Ti3C2 MXene nanosheets for tumor targeting photo- [281] A.V. Kachynski, A. Pliss, A.N. Kuzmin, T.Y. Ohulchanskyy, A. Baev, J. Qu,
thermal/photodynamic/chemo synergistic therapy, ACS Appl. Mater. Interfaces 9 P.N. Prasad, Photodynamic therapy by in situ nonlinear photon conversion, Nat.
(2017) 40077–40086, https://doi.org/10.1021/acsami.7b13421. Photon. 8 (2014) 455–461, https://doi.org/10.1038/nphoton.2014.90.
[258] S.H. Li, W. Yang, Y. Liu, X.R. Song, R. Liu, G. Chen, C.H. Lu, H.H. Yang, [282] F. Li, Y. Du, J. Liu, H. Sun, J. Wang, R. Li, D. Kim, T. Hyeon, D. Ling, Responsive
Engineering of tungsten carbide nanoparticles for imaging-guided single 1,064 nm assembly of upconversion nanoparticles for pH-activated and near-infrared-trig-
laser-activated dual-type photodynamic and photothermal therapy of cancer, gered photodynamic therapy of deep tumors, Adv. Mater. 30 (2018) 1802808,
Nano Res. 11 (2018) 4859–4873, https://doi.org/10.1007/s12274-018-2075-z. https://doi.org/10.1002/adma.201802808.
[259] A.R. Hilgenbrink, P.S. Low, Folate receptor-mediated drug targeting: from ther- [283] S. Wang, L. Zhang, C. Dong, L. Su, H. Wang, J. Chang, Smart pH-responsive up-
apeutics to diagnostics, J. Pharmacol. Sci. 94 (2005) 2135–2146, https://doi.org/ conversion nanoparticles for enhanced tumor cellular internalization and near-
10.1002/jps.20457. infrared light-triggered photodynamic therapy, Chem. Commun. 51 (2015)
[260] S.D. Weitman, R.H. Lark, L.R. Coney, D.W. Fort, V. Frasca, V.R. Zurawski, 406–408, https://doi.org/10.1039/C4CC08178A.
B.A. Kamen, Distribution of the folate receptor GP38 in normal and malignant cell [284] W. Wang, L. Lin, X. Ma, B. Wang, S. Liu, X. Yan, S. Li, H. Tian, X. Yu, Light-induced
lines and tissues, Canc. Res. 52 (1992) 3396–3401. hypoxia-triggered living nanocarriers for synergistic cancer therapy, ACS Appl.
[261] W.A. Franklin, M. Waintrub, D. Edwards, K. Christensen, P. Prendegrast, J. Woods, Mater. Interfaces 10 (2018) 19398–19407, https://doi.org/10.1021/acsami.

26
J. Chen, et al. Biomaterials 237 (2020) 119827

8b03506. [296] L. Larue, A. Ben Mihoub, Z. Youssef, L. Colombeau, S. Acherar, J.C. André,
[285] J. Li, X. Meng, J. Deng, D. Lu, X. Zhang, Y. Chen, J. Zhu, A. Fan, D. Ding, D. Kong, P. Arnoux, F. Baros, M. Vermandel, C. Frochot, Using X-rays in photodynamic
Z. Wang, Y. Zhao, Multifunctional micelles dually responsive to hypoxia and therapy: an overview, Photochem. Photobiol. Sci. 17 (2018) 1612–1650, https://
singlet oxygen: enhanced photodynamic therapy via interactively triggered pho- doi.org/10.1039/C8PP00112J.
tosensitizer delivery, ACS Appl. Mater. Interfaces 10 (2018) 17117–17128, [297] X.-D. Ren, X.-Y. Hao, H.-C. Li, M.-R. Ke, B.-Y. Zheng, J.-D. Huang, Progress in the
https://doi.org/10.1021/acsami.8b06299. development of nanosensitizers for X-ray-induced photodynamic therapy, Drug
[286] F. Moret, E. Reddi, Strategies for optimizing the delivery to tumors of macrocyclic Discov. Today 23 (2018) 1791–1800, https://doi.org/10.1016/J.DRUDIS.2018.
photosensitizers used in photodynamic therapy (PDT), J. Porphyr. 05.029.
Phthalocyanines 21 (2017) 239–256, https://doi.org/10.1142/ [298] L. Song, P.-P. Li, W. Yang, X.-H. Lin, H. Liang, X.-F. Chen, G. Liu, J. Li, H.-H. Yang,
S1088424617300014. Low-dose X-ray activation of W(VI)-Doped persistent luminescence nanoparticles
[287] H.-R. Jia, Y.-W. Jiang, Y.-X. Zhu, Y.-H. Li, H.-Y. Wang, X. Han, Z.-W. Yu, N. Gu, for deep-tissue photodynamic therapy, Adv. Funct. Mater. 28 (2018) 1707496,
P. Liu, Z. Chen, F.-G. Wu, Plasma membrane activatable polymeric nanother- https://doi.org/10.1002/adfm.201707496.
anostics with self-enhanced light-triggered photosensitizer cellular influx for [299] H. Chen, G.D. Wang, Y.-J. Chuang, Z. Zhen, X. Chen, P. Biddinger, Z. Hao, F. Liu,
photodynamic cancer therapy, J. Contr. Release 255 (2017) 231–241, https://doi. B. Shen, Z. Pan, J. Xie, Nanoscintillator-mediated X-ray inducible photodynamic
org/10.1016/J.JCONREL.2017.04.030. therapy for in vivo cancer treatment, Nano Lett. 15 (2015) 2249–2256, https://
[288] P. Wang, X. Li, C. Yao, W. Wang, M. Zhao, A.M. El-Toni, F. Zhang, Orthogonal doi.org/10.1021/nl504044p.
near-infrared upconversion co-regulated site-specific O2 delivery and photo- [300] T.K. Pandita, S. Pandita, S.R. Bhaumik, Molecular parameters of hyperthermia for
dynamic therapy for hypoxia tumor by using red blood cell microcarriers, radiosensitization, Crit. Rev. Eukaryot. Gene Expr. 19 (2009) 235–251, https://
Biomaterials 125 (2017) 90–100, https://doi.org/10.1016/J.BIOMATERIALS. doi.org/10.1615/CritRevEukarGeneExpr.v19.i3.50.
2017.02.017. [301] M.R. Horsman, J. Overgaard, Hyperthermia: a potent enhancer of radiotherapy,
[289] A. Raza, U. Hayat, T. Rasheed, M. Bilal, H.M.N. Iqbal, “Smart” materials-based Clin. Oncol. 19 (2007) 418–426, https://doi.org/10.1016/J.CLON.2007.03.015.
near-infrared light-responsive drug delivery systems for cancer treatment: a re- [302] M. Yao, L. Ma, L. Li, J. Zhang, R. Lim, W. Chen, Y. Zhang, A new modality for
view, J. Mater. Res. Technol. 8 (2019) 1497–1509, https://doi.org/10.1016/J. cancer treatment-nanoparticle mediated microwave induced photodynamic
JMRT.2018.03.007. therapy, J. Biomed. Nanotechnol. 12 (2016) 1835–1851 http://www.ncbi.nlm.
[290] D.K. Chatterjee, L.S. Fong, Y. Zhang, Nanoparticles in photodynamic therapy: an nih.gov/pubmed/29359896.
emerging paradigm, Adv. Drug Deliv. Rev. 60 (2008) 1627–1637, https://doi.org/ [303] H.A. Lankes, C.N. Zanghi, K. Santos, C. Capella, C.M.P. Duke, S. Dewhurst, In vivo
10.1016/J.ADDR.2008.08.003. gene delivery and expression by bacteriophage lambda vectors, J. Appl. Microbiol.
[291] M. Yang, T. Yang, C. Mao, Enhancement of photodynamic cancer therapy by 102 (2007) 1337–1349, https://doi.org/10.1111/j.1365-2672.2006.03182.x.
physical and chemical factors, Angew. Chem. Int. Ed. 58 (2019) 14066–14080, [304] G. Abbineni, S. Modali, B. Safiejko-Mroczka, V.A. Petrenko, C. Mao, Evolutionary
https://doi.org/10.1002/anie.201814098. selection of new breast cancer cell-targeting peptides and phages with the cell-
[292] S. Clement, W. Chen, W. Deng, E.M. Goldys, X-ray radiation-induced and targeted targeting peptides fully displayed on the major coat and their effects on actin
photodynamic therapy with folic acid-conjugated biodegradable nanoconstructs, dynamics during cell internalization, Mol. Pharm. 7 (2010) 1629–1642, https://
Int. J. Nanomed. 13 (2018) 3553–3570, https://doi.org/10.2147/IJN.S164967. doi.org/10.1021/mp100335s.
[293] K. Kirakci, J. Zelenka, M. Rumlová, J. Martinčík, M. Nikl, T. Ruml, K. Lang, [305] N. Gandra, G. Abbineni, X. Qu, Y. Huai, L. Wang, C. Mao, Bacteriophage biona-
Octahedral molybdenum clusters as radiosensitizers for X-ray induced photo- nowire as a carrier for both cancer-targeting peptides and photosensitizers and its
dynamic therapy, J. Mater. Chem. B. 6 (2018) 4301–4307, https://doi.org/10. use in selective cancer cell killing by photodynamic therapy, Small 9 (2013)
1039/C8TB00893K. 215–221, https://doi.org/10.1002/smll.201202090.
[294] H. Wang, B. Lv, Z. Tang, M. Zhang, W. Ge, Y. Liu, X. He, K. Zhao, X. Zheng, M. He, [306] B. Cao, M. Yang, Y. Zhu, X. Qu, C. Mao, Stem cells loaded with nanoparticles as a
W. Bu, Scintillator-based nanohybrids with sacrificial electron prodrug for en- drug carrier for in vivo breast cancer therapy, Adv. Mater. 26 (2014) 4627–4631,
hanced X-ray-induced photodynamic therapy, Nano Lett. 18 (2018) 5768–5774, https://doi.org/10.1002/adma.201401550.
https://doi.org/10.1021/acs.nanolett.8b02409. [307] X. Li, C. yoon Kim, J.M. Shin, D. Lee, G. Kim, H.M. Chung, K.S. Hong, J. Yoon,
[295] C.-C. Hsu, S.-L. Lin, C.A. Chang, Lanthanide-doped core–shell–shell nanocompo- Mesenchymal stem cell-driven activatable photosensitizers for precision photo-
site for dual photodynamic therapy and luminescence imaging by a single X-ray dynamic oncotherapy, Biomaterials 187 (2018) 18–26, https://doi.org/10.1016/j.
excitation source, ACS Appl. Mater. Interfaces 10 (2018) 7859–7870, https://doi. biomaterials.2018.09.041.
org/10.1021/acsami.8b00015.

27

You might also like