Sunteți pe pagina 1din 54

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/271137925

Nanoparticles in Photodynamic Therapy

Article  in  Chemical Reviews · January 2015


DOI: 10.1021/cr5004198

CITATIONS READS

769 417

3 authors, including:

Sasidharan S Lucky
Institute of Bioengineering and Nanotechnology
12 PUBLICATIONS   1,219 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Establishment of In vitro 3D cultures from patient derived xenograft models for drug screening and disease modelling View project

All content following this page was uploaded by Sasidharan S Lucky on 07 January 2019.

The user has requested enhancement of the downloaded file.


Review

pubs.acs.org/CR

Nanoparticles in Photodynamic Therapy


Sasidharan Swarnalatha Lucky,†,§ Khee Chee Soo,‡ and Yong Zhang*,†,§,∥

NUS Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, Singapore, Singapore
117456

Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore 169610
§
Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore 117576

College of Chemistry and Life Sciences, Zhejiang Normal University, Zhejiang, P. R. China 321004
Notes AU
Biographies AU
Acknowledgments AV
References AV

1. INTRODUCTION: PHOTODYNAMIC THERAPY FOR


CANCER
Cancer accounted for 7.6 million deaths (around 13% of all
deaths) worldwide in 2008.1 Almost 13 million cancer cases are
newly diagnosed every year, and deaths are projected to rise,
with an estimated 13.1 million in 2030. Although regular
screening and surveillance programs as well as early
CONTENTS
intervention are the best ways to improve the outcome and
1. Introduction: Photodynamic Therapy for Cancer A survival, efforts need to be divided at finding better cancer
2. Principle of Photodynamic Therapy B therapeutic options that are effective, efficient, affordable, and
2.1. Photodynamic Reaction B acceptable to patients. The conventional cancer treatment
2.2. Mechanism of Tumor Destruction B options are chemotherapy, radiotherapy, and surgery, and more
2.2.1. Direct Tumor Cell Kill B recently small molecule-based therapies and immunotherapy
2.2.2. Vascular Damage D along with a combination of these strategies are being practised.
2.2.3. Inflammatory and Immune Response E However, chemotherapy is often associated with systemic side-
3. Photosensitizers F effects, high recurrence rate is associated with surgical resection
3.1. 1st Generation Photosensitizers F of tumors, while radiation therapy is limited by the cumulative
3.2. 2nd Generation Photosensitizers F radiation dose. While refinement of the conventional cancer
3.3. 3rd Generation Photosensitizers G treatment modalities is important, research has also focused on
4. Current Limitations of Photodynamic Therapy G developing alternate treatment modalities that are safe, potent,
4.1. Tumor Selectivity H and cost-effective. Photodynamic therapy (PDT) is an
4.2. Formulation of Photosensitizers H alternative tumor-ablative and function-sparing oncologic
4.3. Tissue Penetration Depth H intervention. Since its inception in early 1900s and its first
5. Nanoparticles in Photodynamic Therapy I modern demonstration by Dougherty et al. in 1975,2 PDT has
5.1. Nanoparticles As Delivery Vehicles of Photo- undergone extensive investigations and has emerged as a
sensitizers I disease site specific treatment modality. Essentially, it involves
5.1.1. Biodegradable Nanoparticles I the administration of a tumor-localizing photosensitizer (PS)
5.1.2. Nonbiodegradable Nanoparticles P followed by local illumination of the tumor with light of a
5.2. Nanoparticles as Downconverting Photo- specific wavelength to activate the PS. The excited PS then
sensitizers V transfers its energy to molecular oxygen, thus generating
5.2.1. Fullerenes V cytotoxic reactive oxygen species (ROS), such as singlet oxygen
5.2.2. Titanium Dioxide Nanoparticle W (1O2) that can oxidize key cellular macromolecules leading to
5.2.3. Zinc Oxide Nanoparticle W tumor cell ablation.3 Hence, PDT employs 3 nontoxic
5.3. Nanoparticles as Energy Transducers X components that by its own do not have any toxic effects on
5.3.1. X-ray Activatable Nanoparticle X the biological systems, unlike chemotherapy drugs that induce
5.3.2. Quantum Dots Y systemic toxicity and ionizing light of radiation therapy that
5.3.3. Two-Photon Absorbing Nanoparticles Y damages neighboring normal tissues. Clearly, PDT has its own
5.3.4. Upconversion Nanoparticles AA merits compared to the conventional treatment methods due to
6. Conclusion and Future Perspectives AS
Author Information AU Received: August 3, 2014
Corresponding Author AU

© XXXX American Chemical Society A DOI: 10.1021/cr5004198


Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 1. Schematic illustration of a typical photodynamic reaction. Adapted from ref 10. 2002, Elsevier.

its minimal invasiveness, repeatability without cumulative PS can then directly interact with a substrate, such as the cell
toxicity, excellent functional and cosmetic results, reduced membrane or a molecule, and transfer a proton or an electron
long-term morbidity, and improved quality of life of the to form a radical anion or cation, respectively, which then reacts
patients. Over the last four decades PDT has proven to be with oxygen to produce oxygenated products such as
effective in superficial bladder cancer,4 early and obstructive superoxide anion radicals, hydroxyl radicals, and hydrogen
lung cancer,5 Barrett’s esophagus,6 head and neck cancers,7 and peroxides (type I reaction). Alternatively, the energy of the
skin cancer.8 It is also being used as an adjunctive therapy excited PS can be directly transferred to molecular oxygen
following surgical resection of tumor, to reduce residual tumor (itself a triplet in the ground state), to form 1O2 (type II
burden.9 reaction). The energy required for the transition of oxygen
Despite the widespread and rapidly growing applications, from triplet ground state to excited singlet state is 22 kcal
PDT has yet to gain clinical acceptance as a first-line mol−1, which corresponds to a wavelength of 1274 nm.12 Thus,
oncological intervention due to certain limitations including a relatively small energy is only needed to produce 1O2.13 The
lack of an ideal PS, challenges in formulating PS, choosing the byproducts formed as a result of the type I and type II reactions
right light dosimetry for a complete and effective treatment, are responsible for the cell-killing and therapeutic effect in
difficulties in planning the treatment and monitoring the PDT. It is to be noted that both type I and type II reactions can
treatment response; which will be discussed in detail in this occur simultaneously, and the ratio between these processes
review. The application of nanoparticles in PDT has been a depends on the type of PS, as well as the concentrations of
major stride forward in resolving some of the challenges molecular oxygen and substrate present.11 However, most of
associated with classic PS. Since a comprehensive review, the studies indicate type II reactions, hence 1O2 play a
encompassing the theory behind the design of nanoconstruct to dominant role in PDT.14
its application in PDT, is beyond the scope of this review, we 2.2. Mechanism of Tumor Destruction
have attempted to provide an overview of the present status
Following PDT, the extent of photoinduced tumor destruction
and prospects of such nanoparticles by highlighting its
depends on various factors such as the type of PS, its
development in each phase and giving special emphasis on
concentration and localization in the tumor microenvironment
multifunctional theranostic agents, by taking specific illustra-
or its subcellular location at the time of irradiation, the time
tions from recently published articles. The examples given in
between PS administration and light irradiation (drug-light
this review do not mean that the pioneering contributions
interval, DLI), light fluence rate, the total fluence, the type of
made by a large number of researchers are neglected. Special
tumor, and its level of oxygenation.3a,15 PDT’s downstream
interest is devoted to the last section on “Upconversion
targets include tumor cells, tumor as well as normal
Nanoparticles” (UCNs), a multifaceted tool that due to its
microvasculature, and the inflammatory and immune host
recent accelerated progress shows great potential in augmenting system. The ROS generated by the photodynamic reaction is
the scope of PDT in the treatment of solid tumors. For detailed the key effector causing irreversible damage to the tumor cells
information on other nanoparticles, readers are requested to and microvasculature, ensuing a plethora of inflammatory and
refer to key publications and review articles provided in each immune response; a combination of which often helps to
section. achieve a long-term tumor control.15b
2.2.1. Direct Tumor Cell Kill. Generally, the site of
2. PRINCIPLE OF PHOTODYNAMIC THERAPY photodamage is considered to coincide with the location of
accumulation of the PS in the tumor cells, as 1O2 has a very
2.1. Photodynamic Reaction short life (half-life: 0.03 to 0.18 ms),16 restricting its diffusion in
A PS absorbs a photon of light and becomes activated from biological systems to a radius of <0.02 μm.17 Oxidation of the
ground state to a short-lived excited singlet state (1PS*; Figure lipids, amino-acids, and proteins by 1O2 induces irreversible
1). The excited PS may either decay back to the ground state by photodamage to vital subcellular targets such as plasma
emitting fluorescence, a property that can be clinically exploited membrane and organelles like mitochondria, lysosomes, Golgi
for imaging and photodetection; or it can undergo intersystem apparatus, and endoplasmic reticulum (ER). The location
crossing whereby the spin of its excited electron inverts to form where the PS ultimately resides within the cell generally
a relatively long-lived triplet state (3PS*).11 The triplet excited depends on its structural features such as the net ionic charge
B DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Table 1. List of 1st and 2nd Photosensitizers That Are Approved for Clinical Use or Are under Clinical Trials

which can range from −4 to +4, the degree of hydrophobicity across the plasma membrane. PSs that are less hydrophobic
expressed as the logarithm of the octanol/water partition tend to be too polar to diffuse across the plasma membrane and
coefficient, as well as the degree of asymmetry present in the PS are therefore taken up by endocytosis.11 As most of the PSs do
molecule.11 Hydrophobic PSs with two or less negative charges not accumulate in cell nuclei, PDT typically has very low
tend to have the highest intracellular uptake and can diffuse chances of causing mutations.3a PDT induced cell death can
C DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 2. Basic structures of porphyrin-based PS. Adapted from ref 27. 2013, MDPI.

occur by apoptosis, necrosis, or autophagy and does not vessels (angiogenesis) to supply them with oxygen and
depend on the phase of the cell cycle.18 Necrotic cell death, also nutrients. Generally, due to poor and incomplete cellular
known as accidental cell death, is often triggered when high PS borders,19 these abnormal and leaky vessels may serve as an
and/or light doses are used. It is typically characterized by additional means for PS accumulation in tumor region as it
swelling of cytoplasm, devastation of organelles, and disruption leaks through. Moreover, PSs bound to carrier molecules such
of the plasma membrane, leading to the release of intracellular as albumin, high-density lipoprotein (HDL), or low-density
contents triggering an inflammatory response. Apoptotic cell lipoprotein (LDL), have high affinity to endothelial cells and
death (programmed cell death) remains the preferred mode of tumor microvascular endothelium because of the presence of
cell death following PDT, which is characterized by cell high number of specific receptors in their structure.20
shrinkage, membrane blebbing, nuclear fragmentation, chro- Activation of PS either confined in the blood circulation,
matin condensation, chromosomal DNA fragmentation, and
localized in endothelial cells, or bound to the vessel walls results
formation of apoptotic bodies that are scavenged by phagocytes
in damage to the endothelial cells characterized by the loss of
in vivo thus preventing an inflammatory response. The mode of
cell death also depends on the location of the PS within the tight junctions between cells and exposure of vascular basement
cell; for example if the PS is localized in the ER or membranes.21 These primary changes within the vessel lumen
mitochondria, the cell death occurs via an apoptotic pathway, leads to the formation of thrombogenic sites and initiates a
while their localization in the plasma membrane or lysosomes cascade of reactions such as platelet aggregation, release of
could delay or block apoptosis. This is when the cell takes vasoactive molecules, leukocyte adhesion, increase in vascular
alternative routes of cell-death, such as the autophagic or permeability, and vessel constriction.22 All these effects
necrotic pathway.18b ultimately lead to partial or complete vascular shutdown,
2.2.2. Vascular Damage. The growth of solid tumors is blood flow stasis and tissue hemorrhages leading to persistent
dependent on their capacity to induce the growth of blood post-PDT tumor hypoxia and long-term tumor control.23
D DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 3. Chemical structures of some 2nd generation PSs. Adapted from ref 27. 2013, MDPI.

2.2.3. Inflammatory and Immune Response. While components such as neutrophils and macrophages which
direct tumor cell kill and vascular damage are mainly converge on the treated site. Macrophages remove the damaged
responsible for initial tumor ablation, antitumor specific
cancer cells by phagocytosis and present proteins from the
immune process and enhancement of host immune system
might play important roles in secondary cytotoxicity which tumor cells to CD4 helper T lymphocytes, which then activate
could probably aid in the long term tumor control and/or CD8 cytotoxic T lymphocytes. The latter then not only induce
complete tumor response to PDT.24 The principal character- necrosis but may also induce apoptosis whenever tumor cells
istic of the inflammatory process is the release of inflammatory are found, leading to long-term tumor control.25 Thus, PDT
mediators that occur from the treated region, which include
has an added advantage of being an immune-stimulatory
components of the complement system, proteinases, perox-
idases, cytokines, growth factors, and other immuno-regu- modality, whereas surgery and chemotherapy are immunosup-
lators.15c,25 This in turn stimulates various white blood cell pressive.
E DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

3. PHOTOSENSITIZERS
Ideally, a PS is expected to have the ability to preferentially
accumulate in tumor tissue and rapidly clear from the normal
tissue. Amphilicity is another must have property for a clinically
successful PS, which when administered systemically will travel
to the target tissue (tumor) unhindered, for which it requires
some degree of hydrophilicity, and then bind to the target cells,
for which it requires some degree of lipophilicity. It must also
have negligible dark-toxicity, high quantum yield of triplet state
formation, and appropriate triplet lifetime to interact with
ground state oxygen or other substrates, to generate a sufficient
amount of ROS. Most importantly, the PS must be activated by
wavelengths of light above 700 nm to allow deeper penetration Figure 4. Chemical structures of some nonporphyrin 2nd generation
of light in tissues. This is because endogenous molecules, like PS. Adapted from ref 27. 2013, MDPI.
hemoglobin, have strong absorption of light below 700 nm,
thus limiting the effective penetration depth of light in target have absorption maxima at wavelengths longer than 630 nm, as
tissues. With so many disparate requirements, it is difficult to well as high extinction coefficients. This category also includes
find or develop any drug that can qualify as an ideal PS. metalated derivatives of PS such as aluminum phthalocyanine
However, many PSs that do not satisfy all these requirements tetrasulfonate (AlPcS4), Si(IV)-naphthalocyanine (SiNC), and
have been approved for clinical use, with some others being tin ethyl etiopurpurin (SnET2), although there is no consistent
tested in clinical trials (Table 1), and they largely belong to the correlation between metalation and increased photodynamic
first and second generation of PSs as discussed in the following activity.28 Generally, the second generation PS presented higher
sections. quantum yields of 1O2, higher tumor-to-normal tissue
3.1. 1st Generation Photosensitizers concentration compared to HpD and, consequently, exhibit a
better antitumor effect. Furthermore, owing to the shorter
Generally, first generation PSs include porphyrin-based PSs
tissue accumulation time of the second generation PS,
that were developed in the 1970s and early 1980s.
treatment time could be reduced as PDT can be carried out
Hematoporphyrin derivative (HpD) (containing a proprietary
on the same day as the administration of the drug. This gives an
mix of porphyrin monomers, dimers, and oligomers) or
opportunity to conduct PDT as an out-patient procedure,
porfimer sodium (the active material in HpD) are examples
making it convenient and accepatable for patients. In addition
of first generation PSs. They were the earliest and most useful
to the rapid treatment time, the second generation PSs
PSs in clinical trials.15c There are several advantages to porfimer
displayed a shorter window of cutaneous photosensitivity (<2
sodium in that it destroys tumors effectively, exhibits negligible
weeks). The properties of the PS largely depends on chemical
dark toxicity, and can be easily formulated in a water-soluble
and physical parameters such as lipophilicity, type and number
preparation for intravenous delivery. However, these PSs have
of charged groups, charge-to-mass ratio, and type and number
relatively weak absorption of light in the red portion of the
of ring and core substituents.29
spectrum where tissue penetration of light is optimal, thus
Although, some second generation PSs such as amino-
significantly reducing the penetration abilities and thus the
levulinic acid (ALA), mono-L-aspartyl chlorin e6 (MACE),
effectiveness of the treatment. Due to their low extinction
Lutexaphyrin, AIPcS4, tetraphenylporphine (TPPS3, TPPS4),
coefficients, administration of a large amount of the drug is
etc. are relatively hydrophilic, most of them especially those
often required to obtain a satisfactory phototherapeutic
composed of porphyrin ring structures such as Chlorin e6
response. Furthermore, DLI is typically 48−72 h, during
(Ce6), meta-tetrahydroxyphenylchlorin (mTHPP), bacterio-
which the patient must be protected from light. Moreover, they
chloropyll-a, 2-[1-hexyloxyethyl]-2-devinylpyropheophorbide-a
readily accumulate and are retained in the normal tissue and
(HPPH), Tookad, SnET2, and unsubstituted phthalocyanines
skin for prolonged period, leading to severe photosensitivity for
are highly hydrophobic. It is well-known that the degree of PS
approximately 4−6 weeks after treatment. But, these side effects
hydrophobicity affects its route of administration as well as the
could be prevented by avoiding sunlight and high-energy light,
biodistribution/pharmacokinetic profile,13,30 and it was re-
or by wearing protective clothing and sunglasses for
ported that hydrophobic PSs exhibited a higher tumor to
approximately 6 weeks after treatment. However, lack of
normal ratio of 7:1 to 8:1, whereas equivalent hydrophilic PS
tumor selectivity, poor bioavailability, and unfavorable bio-
exhibited a mere 2:1 ratio.31 Although, hydrophobic character-
distribution, as well as prolonged photosensitivity was a major
istic could allow the PS to penetrate the cell membrane and
limitation during the initial clinical trials with first generation
locate in the photosensitive subcellular compartment, highly
PSs.26
hydrophobic PS could form aggregates in aqueous solution
3.2. 2nd Generation Photosensitizers particularly under physiological milieu, which could in turn
Porphyrinoid compounds comprising of porphyrin or porphyr- affect its photophysical (1O2 yield) and photokilling properties
in-based macrocyclic structures such as chlorins, bacteriochlor- due to its inadequate pharmacokinetics. Furthermore, hydro-
ins, phthalocyanines, pheophorbides bacteriopheophorbies, phobicity could hamper its solubility in physiological solvents
texaphyrins (Figures 2 and 3), as well as nonporphyrinoid and body fluids, limiting their clinical applications.32 Thus,
compouds like anthraquinones, phenothiazines, xanthenes, clinically successful PS must have balance between the degree
cyanines, and curcuminoids (Figure 4) with known chemical of hydrophilicity and lipophilicity. In order to improve the
structures are collectively known as second generation PSs.27 solubility of these PSs, various hydrophilic polar substituents
They were mostly developed since the late 1980s, to overcome were introduced in the lead PS structure to synthesize
the shortcomings of the first generation PSs. These PSs usually derivatives that were amphiphilic. The porphyrin ring system
F DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

inherently possess 12 positions, which could be substituted control residual tumor regrowth and prevent tumor recurrence
with various groups such as sulfonic acid, carboxylic acid, due to under-illumination of the tumors.38 In some other cases,
hydroxyls, quarternary ammonium salts, carbonyl, pyridinium overillumination could induce excessive toxicity to adjacent
substituents, and so forth, giving countless number of possible normal tissue, again leading to unfavorable response. Unlike
derivatives.33 Apart from the addition of various side groups, radiation therapy, in which the biological response is well
the porphyrin ring system can also be oxidized, extended, and/ correlated with the energy absorbed per unit mass of tissue,
or modified to carry a central ion to alter the photophysical and there is a clear lack of a widely accepted definition of PDT light
pharmacological properties of the molecule. Similar modifica- dose. This is a fairly complicated problem mainly due to the
tions also apply for other second generation PS compounds like inherent complexity of the PDT mechanism along with lack of
perylenequinones, dyes, phenothiazines, anthraquinone deriva- suitable techniques to measure it clinically. Yet, a frequently
tives, etc. In general, a PS containing no charged peripheral practiced, but a rather crude method involves measuring the
groups tends to be more hydrophobic; PSs with three or more light fluence, amount of PS and oxygen, which are then used to
charged substituents are likely to be hydrophilic; and that with calculate the 1O2 yield (explicit dosimetry).39 The distribution
around two charged peripheral groups could probably be of light is determined by the light source characteristics and the
amphiphilic. In addition to this, PS with anionic substituents, tissue optical properties. However, due to the complexity of
such as sulfonate or carboxyl groups, have been reported to tissue architecture and the relatively sparse knowledge on tissue
localize preferentially in the cytoplasm,34 whereas PSs function- optical properties, measurement and interpretation of light
alized with cationic groups traverse the mitochondrial fluence seems rather challenging. Although the distribution of
membrane and accumulate in the mitochondrion.35 However, oxygen and PS can be measured by optical spectroscopy, the
the exact structural characteristics and mechanism behind these actual distribution could vary due to oxygen consumption
specific distributions and localizations are very much unclear, during the photodynamic reaction and photobleaching,
and the question still remains as to how to maximize tumor respectively. Hence, a more dynamic modeling of the
tissue selectivity over normal tissue accumulation. photodynamic process is required. An alternative approach,
3.3. 3rd Generation Photosensitizers involves measuring a “surrogate” directly associated with
biological response (implicit dosimetry).40 For example,
Much of the research in this field now focuses on development measuring the fluorescence photobleaching of the PS, indirectly
of third generation PSs that can be activated with light of a predicts the production of 1O2.40b A third method - direct
longer wavelength, provoke shorter generalized photosensitivity dosimetry, relies on the direct detection of 1O2 itself, either
and more importantly, have better tumor specificity. This can through its own phosphorescence emission or via 1O2-sensitive
be achieved either by (a) modifying existing PS with biologic chromophores.41 Hence, there is a clear need to develop
conjugates such as peptides-/antibody-/antisense-conjugates to integrated dosimetry systems with light delivery devices as well
achieve tumor specific targeting of PSs,36 or (b) chemical as multimodality imaging (anatomic and diffuse optical
conjugation/encapsulation of PS in delivery vehicles or carriers
tomography) systems with real-time PDT response monitoring
that can transport the drug efficiently in blood from the
capabilities, which will enable clinicians to customize and plan
administration site to the target tissue. In short, the third
PDT treatment regimens on a case by case basis.
generation PS will be an improvement of the current second
It is well-known that PDT is an oxygen consuming modality.
generation PSs in terms of their delivery or targeting abilities.
In other words, the antitumor effects of PDT will be severely
Although, many of the third generation PSs have been widely
affected in the absence of tissue oxygen. Tumor hypoxia can
studied and considered to enable selective targeting in vitro,
occur either due to the presence of pre-existing hypoxic cells
very few have been evaluated for clinical applications as their in
that formed as a results of exhaustion of blood supply through
vivo selectivity was not found to be sufficiently high.37
the rapid tumor growth; or could be induced during PDT
through rapid shut down of tumor vasculature and fast
4. CURRENT LIMITATIONS OF PHOTODYNAMIC depletion of local oxygen supply.21 Solid tumors with high
THERAPY fraction of hypoxic cells are reported to have a poor prognosis
Although, PDT has the potential to be a stand-alone modality to PDT, as hypoxic tumor cells in solid tumors are PDT-
for the treatment and management of cancers at different resistant.42 Among the various methods adopted to deal with
stages, currently it is only being used in the treatment of depletion of tissue oxygenation, fractionation of light irradiation
superficial and flat lesions, lesions that are accessible through into controlled light and dark periods,43 as well as reducing the
endoscopes or as a surgical adjuvant and prophylactic treatment fluence rate to 20 mW cm−2,44 has been extensively studied.
of dysplasias and primary lesions. The fundamental problem These techniques are considered to promote reperfusion of
lies in the inability of PDT to treat solid, bulky tumors or deep- tissue oxygen, to compensate for oxygen depletion. However,
seated tumors. It also cannot be used for the treatment of the hypoxia caused by pre-existing hypoxic cells is not affected
advanced disseminated disease because whole body irradiation by these techniques.45 Besides, both reduction in fluence rate
is not possible at least with the currently available technologies. and light fractionation would lead to significant increase in time
This is partly due to the short-comings in the properties of the of irradiation required for delivery of specific light dose. A third
PSs that are currently available, and partly due to the method involves hyperoxygenation therapy, which is found to
incomplete knowledge about the light dose to be delivered to compensate tumor hypoxia in both pre-existing hypoxic cells
the tumor without affecting the surrounding normal tissue; and those that are acutely induced by PDT. When C3H mouse
making it difficult for the clinicians to plan and customize the transplanted with mammary carcinoma cells MCa were
treatment. As PDT is a localized treatment, it essentially subjected to hyperoxygenation, up to 80% of the tumors
requires an appropriate dose of light to reach every point in the showed no regrowth after 60 days; whereas in animals that
tumor, for it to be effective in eradicating the tumor. Despite inhaled room air, only 20% did not regrow tumors.45 Following
advanced Monte Carlo calculations, often clinical PDT fails to hyperoxygenation, oxygen was found to reach critical, chronic
G DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

hypoxic cells and also resulted in better tumor response to PDT application of PDT. The areas that require improvement in
due to enhanced direct and secondary tumor cell kill.46 In the terms of clinical PDT are as follows.
molecular level, the effect of hypoxia is mediated by hypoxia- 4.1. Tumor Selectivity
inducible factor-1 (HIF-1),47 a heterodimeric protein, compris-
ing of HIF-1α and HIF-1β subunits; both of which are Due to the generalized photosensitivity associated with most of
transcription factors. While HIF-1α is cytoplasmic protein the earlier generation PSs, patients find it difficult to make
responsive to oxygen tension, HIF-1β is a nuclear protein that lifestyle changes and stay indoors for a prolonged period, which
is constitutively expressed and independent of oxygen level.48 generally prevents them from consenting to PDT. Although
In normoxic cells, HIF-1α is continuously degraded, whereas some photosensitizing drugs are found to be preferentially
under hypoxic conditions it translocates to the nucleus and retained by the tumor cells compared to the surrounding
heterodimerizes with HIF-1β to form active HIF-1 protein. The healthy tissue, the exact mechanisms that drive this process are
HIF-1 protein, in turn binds and activates various hypoxia not fully understood. However, the abnormal physiology of
responsive genes such as vascular endothelial growth factor tumors such as leaky vasculature, poor lymphatic drainage,
receptor (VEGF), other angiogenic growth factors, glycolytic abnormal stromal composition, decreased pH, and increased
enzymes and cytokines, as an adaptive response.47,49 This will number of low density lipoprotein receptors are often
in turn diminish the therapeutic responses by triggering considered to contribute to the selectivity of photosensiti-
angiogenesis and tumor relapse. Several reports have zers.15c Tumor selectivity could be improved by targeted
documented the upregulation of HIF-1α and VEGF following delivery agents that can transport the PS from the site of
PDT as a result of vascular damage and photochemical administration to the tumor tissue, leading to the selective
consumption of oxygen.50 Integrating PDT with antiangiogenic accumulation of the PS in the tumor. In this way, unfavorable
agents, such as Avastin (bevacizumab), Celecoxib, Prinomastat biodistribution can be avoided, improving bioavailability of the
(AG-3340), PI3−K inhihtors, etc. has clearly demonstrated PS. Consequently, undesirable side effects such as prolonged
improved therapeutic efficacy in preclinical studies.50a However, skin photosensitivity and damage to surrounding healthy tissues
blocking one or two molecular pathways at a time might not be can be prevented.
sufficient, as multiple pathways might be involved in the 4.2. Formulation of Photosensitizers
proliferation of tumor cells. Therefore, combination therapy
To a large extent, the efficacy of PDT treatment depends on
that targets multiple molecular pathways at the same time could
the photochemical, photobiological, and pharmacokinetic
be an attractive therapeutic strategy to achieve complete and
properties of the PS. In general most of the potent PSs have
effective tumor response following PDT. Apart from these
extended delocalized aromatic π electron system, a character-
difficulties, lack of an ideal PS continues to be a major
istic property that allows them to absorb light efficiently. At the
roadblock restricting the clinical utility of PDT. In fact, the
same time due to π−π stacking and hydrophobic interactions,
search for an ideal PS, with majority of the characteristics as
they easily form aggregates in aqueous media. On one hand,
summarized in Table 2,26,38,51 still continues even after nearly 4
hydrophobicity could be a desirable characteristic, as
solubilization of the PS in the lipid bilayer of the cell membrane
Table 2. List of Properties Required in a Clinically has been pointed out to be one of the main factors of PS
Successful PS efficacy. On the other hand, this feature leads to lack of
chemical purity for regulatory approval solubility of PS and contributes to its aggregation in aqueous
ease of synthesis for commercial and large-scale production solution.33,52 This affects the bioavailability of the dye and
no degradation upon light activation to form toxic byproducts hampers its ROS generation ability, resulting in suboptimal
synthesis method with minimal batch variations PDT treatment outcome. Hydrophobicity also complicates
chemical stability for transport, storage and reconstitution preparation of pharmaceutical formulations for parenteral
solubility in injectable solvents and body’s fluids administration.10,53 Again this could be solved by formulating
amphilicity for tissue penetration the PS in suitable carrier systems that can transport PS in its
good selectivity toward tumor tissue stable monomeric form without altering its spectroscopic and
clinically useful half-life in tissues functional properties. To be therapeutically useful, a PS
rapid elimination from nontarget tissue, minimizing skin photosensitivity formulation should deliver the PS in a form that can be readily
negligible dark toxicity or systemic toxicity and selectively internalized by hyperproliferative cancer cells, in
high quantum yield of singlet oxygen generation such a way that it facilitates accurate and convenient dosing. At
high molar extinction coefficient the same time, the carrier should be biodegradable, nontoxic or
good photostability must not release toxic degradation products, or must have no
excitable by near-infrared wavelengths (700−1300 nm) immunogenic properties.
multiple administration routes (oral, intravenous, intratumoral, or inhalational) 4.3. Tissue Penetration Depth
Clinical application of PDT for the treatment of solid or deep-
seated tumors has been hampered by the tissue penetration
decades of PDT. Although many first and second generation depth limit of the visible light required by most of the
PSs have been effective in establishing the viability and conventional PSs for its activation. The typical penetration
feasibility of this technique, it has not been optimum in depth of red light in living tissue used for PDT is only 1−3 mm.
terms of effectiveness. This is because PSs developed for PDT Hence, even if the surface of the target area is illuminated
seems to have been driven chemically, rather than clinically. evenly, the energy of the incident light will fall dramatically at
Focus is needed on solving the problems of the PS or increasing depths below the surface (or at increasing distances
modifiying the existing PS to overcome the 3 major problems within tissue from an interstitial fiber). This is because most
that might be the potential impediment to the clinical tissue chromophores (hemoglobin, melanin, fat etc.) absorb
H DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

light strongly in the visible spectrum.54 Thus, effectiveness of tion, pharmacokinetics, cell uptake, and targeting
PDT greatly decrease with tissue thickness due to strong abilities.32a,65
attenuation of visible light with increasing tissue depth, leading f. They can be designed as multifunctional nanoplatforms
to incomplete treatment and tumor relapse.55 Apparently, for that carry multiple components, for example, imaging
deep penetration of light into tissues, wavelength of the
agents, chemo-drugs, targeting ligands, and “cloaking”
irradiated beam should be in the near-infrared (NIR) region
between 700−1300 nm, which is known as the “optical agents (agents that avoid interference with the immune
window” of biological tissue.56 Although some efforts have been system).66
made to utilize PSs that absorb in the NIR range to improve the In recent years, there have been several reviews that
tissue penetrability,57 the low energy associated with the NIR highlighted the various approaches for utilizing nanoparticles
light at the single-photon level does not fulfill the energy for PDT, which generally categorized nanoparticles into passive
requirement to efficiently excite PSs for sufficient ROS and active nanoparticles either depending on the absence or
generation. This underscores the need of an efficient energy presence of any targeting moieties on their surface, or based on
or light transducers that can absorb light in the NIR region and their involvement in the excitation of PS.32a,66,67 Here, we have
emit in the visible region, which could then activate the PS broadly classified the nanoparticles utilized in PDT based on
near-by. the functions or tasks they perform into three: carriers of PSs,
Different strategies have evolved to overcome the current PSs by itself and energy transducers of PS. We have further
shortcomings with regard to the issues associated with the PS, attempted to provide comprehensive information on the
often by means of a delivery vehicle that enables a stable design, advantages, perspectives and challenges of each of the
dispersion of PS in aqueous systems. Despite enhanced drug nanoformulations along with a critical discussion in order to aid
loading and improved tumor uptake over free drugs, the the future research in the field of photodynamic nanomedicine.
traditional delivery systems such as oil-based drug formulations 5.1. Nanoparticles As Delivery Vehicles of Photosensitizers
(micellar systems) using low molecular weight surfactants such The first report on nanoparticles as delivery vehicles in 1976
as the polyoxyethylated castor oils (e.g., polysorbate 80 focused on the development of nanoparticle-based vaccines for
(Tween-80), Cremophor- EL etc.) have been reported to elicit controlled and slow release of the vaccine, leading to a better
acute hypersensitivity (anaphylactic) reactions in vivo.58 Hence, immune response.68 This marked a significant technological
more stable, biocompatible and tumor selective carrier systems and medical breakthrough, as until then it was considered
are required for efficient delivery of PS. impossible to administer pharmaceutical suspensions (solid
particles dispersed in a liquid) intravenously, due to the obvious
5. NANOPARTICLES IN PHOTODYNAMIC THERAPY risks of embolism. Today, the development of nanoparticle
Nanoparticles represent an emerging technology in the field of based delivery vehicles has made it possible to deliver many
PDT that can overcome most of the limitations of classic PS.59 chemotherapeutic drugs of low therapeutic index, such as
Typically, they are defined as submicroscopic particles between doxorubicin (DOX) and paclitaxel, in patients by selectively
1 and 100 nm in size. Nanoparticles are designed out of a directing them toward the tumors cells. Currently, there are
variety of naturally occurring or synthetic materials and can be numerous nanoformulations that have been either approved or
engineered to carry multiple theranostic agents, in a targeted under clinical trials for various oncological indications.
manner. Depending on the type of nanocarrier and the mode of In 1991, Labib et al. was the first to report the use of cyano-
attachment or loading of PS in it, the use of nanoparticles in acrylic nanospheres (150 to 250 nm) or nanocapsules (10−380
conjunction with PDT could have at least some of the following nm) synthesized by interfacial polymerization in an oil-in-water
advantages: emulsion, encapsulating phthalocyanine or naphthalocyanine
a. They have large surface to volume ratios,60 thus could derivatives.69 The immediate and subsequent studies focused
effectively increase the amount of PS that can be mainly on the development of a sensitizer-delivery system
delivered to the target cells.61 based on such biodegradable polymeric nanoparticles. Unlike
chemotherapeutic drugs, that need to be released from the
b. Nanoparticles may prevent the premature release of PS
carrier to exert its effect on the target cells, the PS need not be
and potential inactivation of the drug by plasma
released from the carrier as long as molecular oxygen and 1O2,
components,62 thereby preventing its nonspecific accu- is able to diffuse in and out of the carrier. Due to this fact, two
mulation in normal tissues, and reducing overall different strategies are often adopted to develop nanoparticle
photosensitivity. carriers for PDT: a) biodegradable nanoparticles (natural or
c. They bestow amphilicity to PS, a property that some synthetic polymer-based nanoparticles) carrying the PS pay-
potentially outstanding PS agents lack. This allows load, which could undergo enzymatic or hydrolytic degradation
nanoparticles to travel unhindered through the blood- and thereby excreted out, minimizing long-term accumulation
stream carrying the PS payload and localize in the tumor of carriers in biological system; or b) nonbiodegradable
tissue.63 nanoparticles (ceramic- or metal-based nanoparticles), which
d. They generally take advantage of the enhanced does not readily degrade in the biological system, but quite
permeability and retention (EPR) effect, a phenomenon often offer in-built multiple functionalities that make them
caused by the abnormal and leaky tumor neovasculature valuable as theranostic agents.”
and poor lymphatic drainage of the tumor tissue, 5.1.1. Biodegradable Nanoparticles. 5.1.1.1. Polyester
facilitating both diffusion of PS carriers into and their and Polyacrylamide Based Nanoparticles. Biopolyesters are
retention within tumor tissue.64 biocompatible polymers that can be naturally occurring such as
e. Their surface can be further modified with functional poly(hydroxyalkanoates) (PHAs) or synthetic polymers like
groups or targeting agents for altering its biological or poly(orthoesters), poly(β-amino esters) (PbAE), as well as
physical properties thereby improving the biodistribu- poly(α-hydroxy esters); the latter which includes poly(D,L-
I DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 5. General PS loading approaches in biodegradable polymeric nanoparticles.

Figure 6. Conceptual illustration of the mechanism of bioreducible nanoparticles for the switchable photoactivity of PpA. The nanoparticles do not
exhibit photoactivity during blood circulation. However, when internalized by cancer cells, the intracellular reduction-triggered cleavage of the
disulfide bonds accelerates the dissociation of the polymeric nanoparticle, causing the rapid release of PpA and restoration of its photoactivity.
Reproduced with permission from ref 79. 2014 American Chemical Society.

lactide) (PLA), poly(glycolide) (PGA), and poly(ε-caprolac- modification of its surface with a variety of biomolecules for
tone) (PCL) are the most widely employed polymers for targeted PS delivery.
micelle and nanoparticle preparation. Earlier reports using such The initial success of using polymeric micelles and
polyester-based (PLA) colloidal delivery systems of PS nanoparticles for the delivery of PS along with good PDT
exhibited greater photodynamic efficiency and reduced photo- outcome saw a surge in the number of articles involving
sensitivity compared to surfactant based carrier systems.10 research in this field, demonstrating how their targeting ability
Encapsulation of PSs within these systems (Figure 5) can and pharmacokinetic functions in vivo are controlled by its
substantially increase its solubility/dispersibility in aqueous composition, size, surface charge, morphology and hydro-
media, thus improving their pharmacokinetic properties. phobicity.70 Most of the second generation PSs have been
Further advantages of using biodegradable polymer based tested out in combination with such polymeric nanoparticles
nanoparticles is the ability to deliver a reasonably good payload especially poly(D,L-lactic-co-glycolic acid) (PLGA),71 a biode-
of the multiple agents (therapeutic PS and diagnostic agents) to gradable copolymer of PLA and PGA, that has been approved
the tumor, shielding it from premature leakage in the blood by FDA for use in humans.72 Such studies on PLGA
thereby maintaining the photostability of photoactive agents nanoparticles encapsulating different PSs have shown varying
and fluorophores. In addition, it offers the possibility of further degree of success in terms of improved photodynamic activity
J DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 7. (A) Schematic of fabrication of SPION encapsulated nanoparticle following a LbL deposition of PAH and DP and removal of the
polystyrene (PS) template. (B) TEM images of SPIONs⊂NC3. (C) T2-weighted MR images with various concentration of contained Fe. (D)
Fluorescence images of ROS generation in HeLa cells treated with SPIONs⊂NC3 and light irradiation. (E) Quantification of induced ROS.
Reproduced with permission from ref 83. 2014 American Chemical Society.

and dramatic tumor-inhibiting effect, enhanced 1O2 production been shown to have enhanced circulation time and remain in
and increased plasma circulation time offering the possibility of the blood compared to bare PLGA nanoparticles.75 For
reducing the amount of PS administered as well as shortening example, PLGA (poly(lactide-co-glycolide)) nanoparticles with
the DLI. A detailed review by Bechet et al.66 and Chatterjee et sizes above 100 nm, there occurs a very rapid elimination from
al.67b in 2008 and book chapter by Lee and Kopelman73 in the blood with the half-life ranging from 13 to 35s, whereas
2011, outlined each stage in the development of these PEGylated PLGA had much longer half-life of close to 7 h.76
nanoconstructs and further discussed its stability, photo- Generally, nanoformulations accumulate within the tumor
cytotoxicity, biodistribution and PDT efficiency. Degradation stroma via EPR effect. However, they can re-enter the
of the polymeric nanoparticle is generally believed to occur via bloodstream via diffusion, resulting in decreased drug
a hydrolytic process, although the possibility of an enzyme- concentration at the target tissue over time. Intracellular uptake
catalyzed degradation has also been considered.74 Once of these nanoconstructs from the extracellular space within the
degraded to biocompatible nontoxic products, they are tumor requires time-resolved cell membrane-mediated pro-
removed from the body by physiological pathways and cesses. Active targeting of nanoparticles to cancer cell-specific
clearance mechanisms. The rate of degradation can be receptor could promote rapid intracellular uptake of the EPR-
modulated by changing the composition, molecular weight accumulated nanoformulations. Along these lines, Master et al.
and ratio of the copolymeric system. Systemic use of such synthesized epidermal growth factor receptor (EGFR) targeting
synthetic polymeric nanoparticles has been limited due to their GE11 peptide-decorated PEG−PCL micelles encapsulating the
rapid opsonisation and removal from the systemic circulation PS Pc4, for rapid intracellular uptake and enhanced PDT of
by the macrophage cells of the mononuclear phagocytic system EGFR-overexpressing A431 human epidermoid carcinoma
(MPS). In order to evade the recognition and removal by the cells.77 Premature biodegradation of the polymers while still
MPS, the nanoparticles are often further surface modified by in circulation could lead to PS leakage from the nano-
polyethylene glycol (PEG) or poly(ethylene oxide) to confer formulation, again resulting in decreased bioavailability in the
“stealth properties”.70e PEGylated PLGA nanoparticles have target tissue. To tackle this problem, bioresponsive polymeric
K DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

systems that make use of the difference between the normal onto a sacrificial template (polystyrene nanoparticles)
and tumor physiology could be utilized. Peng et al. utilized the embedded with superparamagnetic iron oxide nanoparticles
pH differences between the normal (pH remains constant at (SPIONs), followed by dissolution of the template (Figure 7),
7.4) and tumor (pH below 7.2) tissue, to develop 2- was reported by Yoon et al.83 The multifunctional hollow
(diisopropylamino)ethyl methacrylate (DPA) based poly- nanoconstruct showed potential in MR imaging and PDT and
(ethylene glycol) methacrylate-co-DPA (PEGMA-co-DPA) could further be utilized to carry additional therapeutic tools
copolymers that self-assembled to form pH sensitive nano- with in their inner void space.
particles in an aqueous environment, encapsulating hydro- Xiang et al. developed polymeric nanovesicle poly(ethylene
phobic m-THPC.78 The nanoparticles were found to be glycol)-block-poly(D,L-lactic acid) [PEG−PDLLA] loaded with
unstable only below a pH of 6.89, thus releasing the PS in a a chemotherapeutic agent, DOX and a lipophilic PS
site-specific manner. Recently, Kim et al. developed a technique hematoporphyrin monomethyl ether (HMME) [PEG−
for bioreducible activation of encapsulated PS Pheophorbide-A PDLLA-DOX-HMME], for combined PDT and chemotherapy,
(PpA) upon intracellular uptake of the polymeric nano- and confirmed that a synergistic effect of both agents led to an
particle.79 Here, 2 PpA molecules were chemically conjugated increased in vivo cell kill in human hepatocellular carcinoma
to one mPEG molecule with biarmed linkages (Figure 6). The (HepG2) cells.84 Yet another approach for triple modal
amphiphilic construct self-assembled to form core−shell- fluorescence, MR, and photoacoustic imaging (PAT)-guided
structured nanoparticles under aqueous conditions, where the combined photothermal (PTT) and PDT, was very recently
hydrophobic PpA remains in a photoinactive state due to demonstrated by Gong et al. utilizing a nanomicellar system.85
fluorescence resonance energy transfer (FRET) mediated Ce6 grafted to an amphiphilic polymer composed of poly-
intramolecular and intermolecular quenching. This property (maleic anhydride-alt-1-octadecene) (C18PMH) and PEG
would minimize deleterious effect due to any 1O2 generation chains (abbreviated as C18PMHPEG-Ce6), was utilized to
during its systemic circulation. The PS payload carrying encapsulate IR825 (an indocyanine green (ICG) analog); for
polymeric construct accumulates in the tumor stroma via the combined PDT and PTT. In addition to being a PDT and
EPR effect. As the nanoconstruct enters the cytosol of the fluorescence imaging agent, Ce6 also serves as a chelating agent
tumor cell that has a higher concentration of glutathione to capture gadolinium-III, enabling T1 -contrast in MRI. Apart
(GSH) than those in extracellular fluids, the thiol responsive from fluorescence and MR imaging the synthesized nano-
disulfide linkers are rapidly cleaved releasing the photoactive PS construct could also offer contrast in PAT imaging due to its
molecule; resulting in enhanced cell-kill. strong NIR absorbance attributed from IR825. Combined PDT
Of late, multifunctional polymeric nanoparticle based (660 nm light at a power density of 2 mW cm−2 for 1 h; 7.2 J
delivery systems are gaining interest and several studies cm−2) and PTT (808 nm laser at a power density of 0.3 W
reported application of polymeric nanoparticle in carrying cm−2; 108 J cm−2) in vivo, 12 h post intravenous injection of
both diagnostic and therapeutic payload. Gupta et al. reported a IR825@C18PMH−PEG−Ce6-Gd nanomicelles, demonstrated
postloading approach of a NIR excitable cyanine dye (CD) remarkable synergist effect in inhibiting tumor growth. In
fluorophore and red-light absorbing HPPH, at a ratio of 1:2, in conclusion, these nanoparticles present as one of the most
a biodegradable polyacrylamide (PAA) nanoplatform for versatile drug/PS carrying nanoplatforms that could be further
simultaneous tumor-imaging (NIR fluorescence imaging) and manipulated to incorporate a variety of functionalities.
long-term tumor cure by PDT.80 A different approach was 5.1.1.2. Liposomal Nanoparticles. Liposomes are one of the
adopted by Lee et al. where multifunctional PLGA nano- first nanoparticle-based delivery platforms to be applied in
particles (PLGA coupled with methoxy-PEG (mPEG) or Ce6), medicine,86 and there are over 11 liposomal formulations
synthesized by the Steglich esterification method, were utilized approved for clinical use today, with many more in clinical and
to carry an inorganic cargo−iron oxide (Fe3O4).81 The preclinical development. They are concentric phospholipid
synthesized multifunctional PLGA nanoparticles exhibited vesicles consisting of single or multiple bilayered membrane
potential in simultaneous high contrast magnetic resonance structure composed of natural or synthetic lipids (Figure 8A).87
imaging (MRI) and therapy, demonstrating significant Their unique ability to contain hydrophilic drugs in their
regression of tumor volume in human nasopharyngeal aqueous core and hydrophobic agents within their lamellae
epidermal carcinoma-KB tumor-bearing nude mice, following makes them excellent therapeutic carriers.
PDT. Similarly, Wang et al. developed a tumor targeting Various studies have compared the tumor uptake and PDT
multifunctional nanocarrier based on amine-functionalized outcome of liposomal versus nonliposomal PSs under identical
biodegradable PAA nanoparticles, for simultaneous fluores- conditions since the late 1980s. Although liposomes have
cence imaging and PDT in a “see and treat” approach.82 The proved to be biocompatible carriers of PS, they did not emerge
HPPH conjugated PAA nanoparticles were synthesized by a as an ultimate PS delivery system. This is mainly due to three
modified water-in-oil microemulsion method. The presence of reasons. First of all most of the conventional or unmodified
a tumor cell and tumor blood vessel (angiogenic vasculature) liposomes often included cholesterol in its structure to improve
homing F3 peptide, targeted the nanoparticle specifically to the the rigidity of the bilayer membrane. But, the presence of
tumor site. Furthermore, chemical conjugation of CD cholesterol reduced the permeability of encapsulated PS.88
molecules to both surface and inside the matrix of the Second, there exists an exchange of lipids between the
nanoparticles, enabled fluorescence imaging. The synthesized liposomes and HDLs, leading to an irreversible disintegration
tumor-specific nanocarriers containing both fluorophores and of the liposome.89 This causes premature release of the PS in
PS, emitted bright fluorescence and also generated 1O2 upon the bloodstream before reaching the target tissue. Conventional
light irradiation, resulting in an irreversible but selective liposomes are also prone to opsonisation by plasma proteins
destruction of the cancer cells. A novel layer-by-layer (LbL) after which they are quickly removed from the circulation by
deposition of positively charged poly(allylamine hydrochloride) cells of the MPS. Thus, they generally exhibit short plasma half-
(PAH) and negatively charged PS−dendrimer porphyrin (DP) life, reducing the time for tumor cell uptake, hampering their
L DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

ization of the PS, which consequently contributes to increased


tumor accumulation and minimizes the nonspecific photo-
sensitivity of normal tissues. Although, progress has been made
in the comprehension of critical parameters for design and
development of a variety of liposomal formulations of PSs, the
critical parameter to consider when optimizing liposomal PDT
for clinical application such as its pharmacokinetics, biodis-
tribution, drug release, stability of the nanocarrier, and
intratumoral PS localization is still unclear. Reshetov et al.
compared two liposomal formulations of temoporfin, a
conventional liposome (Foslip) and polyethylene glycosylated
liposome (Fospeg), in tumor-grafted mice and found
PEGylated liposomal formulation to be far superior in terms
of blood circulation time, resulting in a rapid and efficient
accumulation of PS in the tumor via EPR effect and thus
significantly shortening the DLI, a characteristic that would
enhance patients interest in PDT.100 On the other hand, Foslip
was soon eliminated from the circulation due to premature
drug release and liposome destruction.
Another area which is gaining popularity is multifunctional
liposomal formulation for theranostic application such as
nanocells101 and the porphysomes.102 While, nanocells are
comprised of PS incorporated polymeric nanoparticles
Figure 8. PS loading strategies in (A) liposome and (B) and (C) encapsulated in a PEGylated liposome, porphysomes are
dendrimer. formed by self-assembly of PS (pyropheophorbide)-phospho-
lipid conjugate into liposome-like structures. In addition to
generalized use as tumoritropic carriers of PSs.90 However, in their drug delivery capabilities, these novel classes of liposomes
the year 2000, a liposomal formulation of PS, labeled as have shown potential in multimodal imaging and therapeutics.
Visudyne, hit the clinics for the treatment of patients with age- Recently, Liang et al. reported yet another multifunctional
related macular degeneration or subfoveal choroidal neo- liposomal carrier named cerasome, developed by chemical
vascularisation secondary to pathological myopia. The lip- conjugation of porphyrin to an organoalkoxysilylated lipid, with
osomal formulation was composed of zwitterionic dimyristoyl- a high drug loading efficiency of 33.46%.103 This is significantly
phosphatidylcholine (DMPC), negatively charged egg phos- higher than that obtained by physically entrapping PS within
phatidylglycerol (EPG), and a hydrophobic PS−Verteporfin or the cerasomes (generally less than 10%). Second, covalent
benzoporphyrin derivative monoacid (BPD-MA), a second linkage could prevent premature release of PSs during systemic
generation PS derived from protoporphyrin IX (PpIX) circulation. The silica-like surface endows the cerasome
dimethyl ester.91 The incorporation of BPD-MA in the remarkably high stability, whereas the inherent porosity allows
liposome enabled its partition into plasma lipoproteins more oxygen molecules to diffuse in and out of the vesicles freely.
rapidly, ensuring higher levels of the PS in the tumor tissues Unlike porphysomes that had almost no photosensitizing effect
and the neovasculature. As liposomal membrane provides as the energy that is normally released for 1O2 generation is
sufficient sites to conjugate multiple functional ligands, dissipated thermally, cerasomes exhibited good fluorescence
liposomes with a specifically modified design, like PEG grafted even at an extremely high number of porphyrins, thus proving
long-circulating liposomes92 or antibody,93 peptide,94 folic acid to be a powerful tool for simultaneous photodynamic
(FA),95 or glycoprotein (transferrin)96 conjugated tumor diagnostics and therapy. However, no studies were performed
targeting liposomes can be designed for enhanced pharmaco- to prove its efficacy in vivo. A different approach by Hasan et al.
kinetic properties as well as improved therapeutics. This has combined the targeting as well as anticancer properties of
been reviewed by Derycke and de Witte89 and more recently by monoclonal antibody Cetuximab (C225), to develop a
Jin and Zheng.97 In a yet another review, Skupin-Mrugalska et liposomal formulation of BPD-MA for simultaneous photo-
al. summarized the modifications of the PS properties in immune/PDT.104 Here, BPD-MA was encapsulated in a
liposomal formulations and further discussed the development, Cetuximab antibody attached preformed plain liposome
preclinical studies, and future perspective of such liposomal (PPL) by physical adsorption. Cetuximab antibody not only
formulations.98 Active targeting of liposome was expected to facilitated selective binding of the nanoconstruct to EGFR
improve the drug delivery and overall PDT efficacy by overexpressing ovarian cancer cells, but also blocked the EGFR
enhancing tumor selectivity of the PS, but the results of these signal transduction pathway, which in combination with PDT
studies are not always very straightforward. In addition, the PS resulted in synergistically enhanced cancer cell destruction.
concentration in liposomes is generally less than 10%. This 5.1.1.3. Dendrimer Based Nanoparticles. Dendrimers are
underscores the need of liposomal vesicles with better stability three-dimensional tree-like branched macromolecules that can
and PS loading efficiency. be tailored in size with functional peripheral groups and inner
Liposomal carriers with triggered-release mechanism, that cavity for incorporation of a variety of molecules. Unlike
selectively release PS upon internal (pH or enzymes) or conventional polymers, dendrimers are prepared by a stepwise
external stimuli (light or temperature), are also gaining more synthetic procedure, leading to a highly ordered branching
attention due to its potential in being truly tumoritropic pattern with consistent and well-defined architecture. Owing to
carriers.99 Such mechanisms seem to enhance cellular internal- its three-dimensional structure and relatively large size, ranging
M DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 9. (A) Synthesis of mono-(PcSi(OH)(mob) 2) and disubstituted (PcSi(mob)2 3) derivatives of the silicon phthalocyanine (PcSi(OH)2 1)
and schematic representation of tumor targeted theranostic platform based on phthalocyanine-loaded dendrimer (Pc−LHRH). (B)
Phototherapeutic effects of Pc−LHRH on A2780/AD cancer cell incubated with different drug concentrations and irradiated for 5 min at 670
nm (36 J cm−2). (C) The influence of laser irradiation time on A2780/AD cells treated with 0.6 μg mL−1 Pc−LHRH and irradiated at 670 nm. (D)
In vivo images of tumor-bearing mouse and ex vivo images of major organs acquired before and 10 h after intravenous injection of Pc−LHRH.
Organs: 1, liver; 2, spleen; 3, kidneys; 4, heart; 5, lungs; 6, tumor. Reproduced with permission from ref 112. 2014 American Chemical Society.

from first up to larger seventh-generation, dendrimers are dendrimer (Figure 8B and C). PS molecules loaded on the
expected to be internalized in membrane-limited organelles, exterior are generally released, either upon photoexcitation and
thereby achieving controlled localization in the intracellular induction of 1O2 that dissolves the covalent linkage between the
compartment.105 Dendrimers have received much attention as a PS and dendrimer108 or by cellular esterases that hydrolyses
PS carrier due to their unique structure, which consists of these bonds.109 Conjugating the PS to the surface of the
several surface groups that can be used to tag different targeting dendrimer serves several advantages: first, it reduces the toxicity
molecules or functional groups. Furthermore, the size and of otherwise toxic dendrimer like polypropyleneimine (PPI);
lipophilicity of the dendrimer-conjugate can be controlled and the PS can be loaded in its monomeric form; prevents
modified for optimizing cellular uptake and tissue biodistribu- premature release of PS and increases the intracellular
tion.106 Thus, the characteristics of the conjugated PS can be accumulation of PS compared to free PS.110 However, the
modulated for optimizing the drug delivery, and thereby photosensitizing activity of the PS could be affected by such
improve the therapeutic efficiency. In their recent review, chemical complexing. Several studies also reported chemical
Klajnert et al. discussed in detail how the application of synthesis of a built in central PS with in the dendrimer core as
dendrimers in PDT can aid in overcoming the obstacles in PS well as PS centered ionic dendrimers capable of forming
delivery to improve PDT outcome.107 The PS can be either polyion micelles through electrostatic interactions with
tagged to the functional groups on the surface of the dendrimer oppositely charged block copolymers.111 However, such
by covalent linkages or encapsulated in the core of the synthesis method often involves long and tedious multistep
N DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

processes required for preparation and purification of the PS- agent DOX following an intratumoral administration of both
core dendrimer. Moreover, this strategy only allows embedding formulations, for a head to head comparison. Although, the PS
of a single PS molecule per dendrimer, resulting in low drug loaded albumin nanoparticles did not induce a significant tumor
loading efficiency. volume reduction, it resulted in a remarkable increase in the
Recently, Taratula et al. reported a simple and efficient necrotic area both in the central region as well as the peripheral
approach to encapsulate hydrophobic silicon-based phthalo- region of the tumor, unlike the other experimental groups in
cyanine (PcSi) into PPI dendrimer (Figure 9).112 The synthesis which the necrosis was localized only to the central part.
step involved modification of the PcSi molecule with a Furthermore, it exhibited very low toxic side effects compared
hydrophobic linker (4-methoxy-4-oxobutanoic acid /(mob)), to treatment with DOX, and thus could be developed in to a
which significantly enhanced physical encapsulation of the PS promising PS delivery agent.
into the hydrophobic pockets of a fourth generation PPI Chitosan. Chitosan is a polysaccharide derived from the
dendrimer with drug encapsulation efficiency of 20% w/w. partial deacetylation of chitin, primarily from crustacean and
Further modification of the complex with PEG and luteinizing insect shells. It consists of repeating units of glucosamine and
hormone-releasing hormone (LHRH) peptide, improved its N-acetyl-glucosamine. Although chitosan is insoluble at neutral
biocompatibility and tumor-targeted delivery. The prepared pH, it is soluble and positively charged at acidic pH.118 Several
formulation required no release of the drug from the carrier, studies have reported the use of amphiphilic chitosan-based
preventing leakage of SiPc into the blood circulation, and at the nanoparticles as carriers of hydrophobic anticancer drugs in
same time exhibited significant ROS production upon laser their inner cores.119 For PDT, the PS was either entrapped in
activation. Furthermore, the nanocarrier-PS conjugate exhibited the inner core of self-assembled chitosan nanoparticles120 or
a distinct NIR absorption (700 nm) and fluorescence emission chemically linked to chitosan which then self-assembles to form
(710 and 815 nm) spectra, required for efficient PDT and a nanoparticle.121 The latter design has given rise to the
fluorescence imaging in the NIR range. possibility of developing “switchable quenching/dequenching”
5.1.1.4. Natural Macromolecule Based Nanoparticles. or “on/off” carrier systems. Such a system remains in a
Various nanoparticles made of cross-linked natural polymers quenched or “off” state with no fluorescence signal and
such as proteins and polysaccharides have also been tested as phototoxicity with light exposure due to FRET based
carriers of PSs. quenching effect of PS within the nanoparticle core. However,
Albumin. Human serum albumin (HSA) based nanoparticles upon cellular uptake the fluorescence signal and 1O2 generation
have attracted interest as theranostic drug carrier particularly was restored due to the cleavage of bonds and release of the
for applications involving lipophilic drugs. This technology PSs from the chitosan nanoparticles. Although, for self-
allows for transportation of hydrophobic drugs without the assembly of chitosan derivatives into nanoparticles, often
need for potential toxic solvents. Moreover, it exploits hydrophobic moieties such as bile acids or fatty acids have to
endogenous albumin pathways for selective delivery of large be conjugated to it. Lee et al. conjugated ursodeoxycholic acid
amounts of chemotherapeutic drugs to the tumor, thus skipping (UDCA), a secondary bile acid, to chitosan (ChitoUDCA) via
the issue of possible accumulation of these drug-nanoparticles EDC-NHS chemistry and incorporated highly anionic and
conjugates in other parts of the body.113 This forms the basis of hydrophobic Ce6 into the cationic UDCA-chitosan nano-
the first U.S. FDA approved nanoparticle-albumin bound particles through ion-complex formation and hydrophobic
paclitaxel (nab-paclitaxel), sold under the trade name of interaction.122 PDT at 664 nm (2 J cm−2) following 2 h
Abraxane, for the treatment of metastatic breast cancer after incubation of Ce6-incorporated ChitoUDCA nanoparticles in
the failure of first line standard chemotherapy. Wacker et al. HuCC-T1 human cholangiocarcinoma cells, enhanced Ce6
employed albumin based nanoparticles as carriers of two uptake in the cells and correspondingly high ROS generation
lipophilic PSs namely, mTHPP and m-tetrahydroxyphenyl- and phototoxicity compared to free Ce6. Recently, Chen et al.
chlorin (mTHPC).114 The PSs were loaded into a preformed coloaded a NIR absorbing PS−ICG and a gold nanorod in
albumin nanoparticle by an absorptive drug loading technique. chitosan nanospheres (CS-AuNR-ICG NSs) prepared by a
Although both formulations generated relatively low amount of nonsolvent counterion complexation method and electrostatic
1
O2 in aqueous solution due to low local oxygen concentration, interaction, for combined PTT/PDT.123 The 180 nm CS-
efficient 1O2 generation and consequently effective cell death AuNR-ICG NSs were designed to be triggered by single NIR
was reported when the PS loaded nanoparticles were incubated laser at 808 nm, allowing efficient heat generation and ROS
with cells. Despite being an efficient carrier for PSs, there are production at the same time. NIR radiation (2 W cm−2 for 10
still a number of problems associated with the development of min) of murine hepatic H22 xenograft tumors in mouse model,
albumin nanoparticles as delivery agents, including complicated 8 h post intravenous injection of CS-AuNR-ICG NSs (7 mg
fabrication methods, wide size distribution, instability under kg−1 Au equivalent plus 6 mg kg−1 ICG equivalent), led to
physiological conditions, and the unintended release of PSs significant reduction in tumor growth and prolonged survival
before it reaches the target site.115 Thereafter, Jeong et al. due to a synergistic PTT/PDT effect.
devised an alternative method of conjugating Ce6 to lysine Gelatin. Gelatin, a water-soluble macromolecule obtained by
residues within HSA so as to form self-assembling nano- partial hydrolysis of porcine or bovine collagen, is a
particles.116 The developed Ce6 bound albumin nanoparticles proteinaceous polymer widely utilized as a food additive.
exerted higher tumor specificity mainly due to the prolonged They make good drug carrier systems because of their
circulation in the blood in addition to the EPR effect and biocompatibility, relatively low antigenicity, and the presence
exhibited better therapeutic efficacy compared with free Ce6 of free amino groups on their surface which can be exploited for
following irradiation. Recently, Portilho et al.117 fabricated an surface modification with target molecules.124 Babu et al.
albumin-based nanosphere containing zinc-phthalocyanine explored the potential of PEG modified-gelatin nanoparticles
tetrasulfonate (ZnPcS4-AN) and compared PDT using this loaded with hypocrellin B (HB) for PDT.125 Despite being an
PS loaded nanoconstruct with standard chemotherapeutic efficient PDT agent, problems like poor drug loading and rapid
O DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

drug release from the gelatin nanoparticles called for further degraded through biochemical decomposition of the silicon−
improvements to this carrier system. These issues were carbon bond128 and presence of the organic group confers
addressed in a subsequent paper by the same group, in which some degree of flexibility to the otherwise rigid silica matrix,
they developed a carrier system based on PEG modified-gelatin which enhances the stability of such particles in aqueous
nanoparticles associated with hydrophobic PLA biopolymers systems. MSNs are typically synthesized by sol−gel processes in
and loaded with hypocrellin B (HB).126 Here, incorporation of the presence of a surfactant such as C12-trimethylammonium
PLA was considered to support enhanced loading of hydro- bromide or C16-trimethylammonium bromide, to control pore
phobic drugs, and thereby the therapeutic efficacy of the sizes.
associated hydrophobic PS. The average size of the nanocarriers In the 2010 review by Couleaud et al., the different method
were comparatively smaller, with relatively better drug loading of synthesis of silica nanoparticles and the two main PS loading
capabilities exhibiting slow release of drug from the nano- strategies (noncovalent and covalent) are discussed in detail.129
particles. Furthermore, the HB loaded nanocariers evoked a Physical entrapment of PS inside the silica network may lead to
superior phototoxicity compared to free-HB in vitro. premature release of the PS from the carrier resulting in
5.1.2. Nonbiodegradable Nanoparticles. Due to their reduced treatment efficiency and side-effects. Covalent coupling
inability to degrade and release drugs in a controlled manner, of the PS inside or at the surface of the nanoparticles is
nonbiodegradable nanoparticles are not traditionally used as a expected to overcome these drawbacks. One of the first reports
drug carrier. However, nonbiodegradable nanoparticles have in this field by Prasad et al. discussed the synthesis of 30 nm
gained much attention in the field of PDT as multifunctional ORMOSIL nanoparticles encapsulating water insoluble
theranostic carriers due to their unique characteristics such as HPPH.130 The entrapped PS was found to generate 1O2
optical properties and tunability in its shape, size, porosity, etc. efficiently upon irradiation, due to the inherent porosity of
They usually include inorganic, metallic, or composite nano- the nanoparticles. The fine pores (0.5−1 nm in diameter) in
particles that may not degrade readily in the biological system the ORMOSIL nanoparticles are small enough to block drug
when compared to polymeric biodegradable nanoparticles. release but permeable to molecular oxygen and the generated
1
While, the latter undergo degradation into biologically benign O2. In a subsequent study by the same group, the PS
components, nonbiodegradable nanoparticles do not degrade iodobenzylpyropheophorbide was covalently incorporated to
readily and do not release the accompanying PS at the time of the ORMOSIL nanoparticles (20 nm), to avoid premature
their activation but allow the generated 1O2 to diffuse out of the release of PS often associated with noncovalent encapsulation
carrier. Thus, these nanoplatforms can be considered as a of PS.131 Again, the incorporated PS molecules retained their
modular system that contains thousands of PS molecules, spectroscopic and functional properties and could generate 1O2
bringing them as a single, nondepleting synergistic unit to the robustly upon photoexcitation. This was followed by a series of
tumor cells. studies using ORMOSIL nanocarriers embedding different PS
5.1.2.1. Silica Nanoparticles. For application in PDT, silica such as phthalocyanine 4 (Pc4),132 PpIX,133 and mTHPC.134
based nanoparticles such as Stö ber silica nanoparticles, Tang et al. compared PS loading and PDT efficiency of
organically modified silica (ORMOSIL), and mesoporous silica hydrophilic methylene blue (MB) in 20−30 nm hydrophilic
nanoparticles (MSN) are frequently used. These nanoparticles polyacrylamide nanoparticles, 190 nm sol−gel silica nano-
are quite promising as vectors for PDT applications due to its particles and 160 nm ORMOSIL nanoparticles.135 As expected,
chemical inertness, transparency of the matrix to light MB loading in the smaller polyacrylamide nanoparticle was
absorption and porosity that is not susceptible to swelling or found to be the lowest. The more spacious and negatively
alterations with a varying pH. They can be prepared using a charged sol−gel nanoparticles demonstrated higher loading of
variety of precursors and many routes of synthesis are available the positively charged MB compared to ORMOSIL nano-
offering great flexibility. The nanoparticles are tunable in terms particles that has a unique internal hydrophilic and external
of their size, shape, porosity, and dispersibility. Furthermore, hydrophobic layer. Although, the Stöber nanoparticles gen-
they can be surface modified with a variety of functional erated a higher amount of 1O2 delivered per milligram of
moieties, polymers such as PEG, or targeting biomolecules for nanoparticle due to their high PS loading efficiency, the
tumor-cell targeting. reaction of the generated 1O2 with ADPA after irradiation at
Spherical and monodisperse silica nanoparticles are generally 650 nm showed a higher kinetic rate with ORMOSIL
prepared following the sol−gel procedure proposed by Stöber nanoparticles than with Stöber method.
et al. in 1968.127 By this method, silica nanoparticles of different MSNs possess large surface area and high accessible pore
sizes ranging from 50 nm to 1 μm can be prepared from volume, making them very suitable as drug delivery agents.
aqueous alcohol solutions of silicon alkoxides in the presence of Qian et al. reported the synthesis of ultrasmall (25 nm) and
ammonia as a catalyst, where the size of particles depends on highly monodispersed MSNs encapsulating PpIX.136 These
the type of silicon alkoxide and alcohol. This method has nanoparticles were efficiently taken up by HeLa cells and
several advantages such as the synthesis may be carried out at caused cell-death via necrotic pathway upon photoexcitation of
low temperature, as well as desired pH to yield high purity PpIX at 532 nm light within 8 min post irradiation. Recently,
products and the reaction kinetics of the process may be targeted delivery of such PS loaded multifunctional MSNs are
controlled by varying the composition of the reaction mixture. gaining interest. Brevet et al. developed mannose-functionalized
ORMOSILs, on the other hand, are prepared from the MSNs encapsulating a water-soluble anionic porphyrin, for
precursor alkoxy organosilane in an oil-in-water microemulsion, cancer cell specific targeting.137 These nanocarriers presented
through a complex purification process. The presence of both much higher photokilling efficiency in MDA-MB-231 cancer
hydrophobic and hydrophilic groups on the precursor helps cells in vitro, through mannose dependent endocytosis of the
them to self-assemble as both normal micelles and reverse functionalized nanocarriers. However, when physically ad-
micelles under appropriate conditions, giving rise to nano- sorbed into the mesoporous structures of MSNs, there is a
particles of about 160 nm in size. ORMOSILs can possibly be high probability of PS molecules being prematurely released
P DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 10. (A) Synthetic process of Ce6-doped mesoporous silica nanorods (CMSNRs). (B) TEM images of CMSNRs (the upper row). Those
samples with different aspect ratios were prepared by adding increasing amount of Ce6. (C) Schematic illustration for of the loading of DOX into
CMSNRs. (D) Relative viabilities of 4T1 cells after incubation with different concentrations of Ce6 and CMSNR4 under 660 nm light irradiation (5
mW cm−2, 1 h). (E) Relative viabilities of 4T1 cells after incubation with various concentrations of free DOX and CMSNRs4/DOX in the dark. (F)
Relative viabilities of 4T1 cells after treated with CMSNRs4 plus light, CMSNRs4/DOX in the dark, and CMSNRs4/DOX plus light (660 nm, 5
mW cm−2, and 1h). P values were calculated by Tukey’s post-test (***p < 0.001, **p < 0.01, or *p < 0.05). (G) Tumor growth curves of different
groups of mice after various treatments indicated (5 mice per group). Error bars were based on standard errors of the mean. Reproduced with
permission from ref 141. 2014 Springer-Verlag Berlin Heidelberg.

from the carrier, leading to a reduced efficiency of PDT. Zhang synthesized a folate conjugated phospholipid-capped PpIX-
et al. developed multifunctional core−shell structured nano- loaded and FITC-sensitized mesoporous silica nanocarriers, for
carrier composed of a nonporous FITC dye-doped silica core targeted intracellular delivery of the nanocarriers in folic-acid
and a mesoporous silica shell containing PS, hematoporphyrin receptor overexpressing HeLa cells.139 The developed nano-
(HP).138 Such a design allowed simultaneous fluorescence construct was found to be superior to free PpIX in terms of its
imaging and PDT. The HP molecules were covalently linked to intracellular delivery efficiency and decreased dark toxicity in
the mesoporous silica shell, exhibited excellent photo-oxidation vitro. In vivo PDT at 630 nm following intratumoral injection
efficiency and cell imaging abilities. However, cell killing of folate tagged nanoconstruct in nude mice bearing B16F10
efficiency following PDT was not tested in vitro. Teng et al. melanoma tumors, caused around 65% destruction of tumors.
Q DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 11. (A) Design of PEG-SiPc4-AuNP. (B) In vivo fluorescence imaging of PEG-SiPc4-AuNP conjugate injected mouse at various time points
within 24 h. Arrows indicate tumor location. (C) Assessment of PEG-AuNP-SiPc4 biodistribution on Au content evaluated by GFAAS. (D)
Histology studies of the organ tissues from mice injected with PEG-AuNP-SiPc4 after 4 h, 24 h, and 7 days postinjection. The black and brownish
spots indicate AuNPs in the tissues. Images were captured at 40× magnification. Scale bar represents 100 μm. (E) fluorescence images of organs
from the injected mice showing biodistribution and elimination of PEG-SiPc4-AuNP based on Pc 4 fluorescence. Reproduced with permission from
ref 146. 2011 American Chemical Society.

Another dual functional nanoconstruct based on mesoporous amount of Ce6 during the synthesis step was found to change
silica composite nanoparticles was fabricated by Zhao et al. for the shape of the MSN from spheres to rods with aspect ratios
the codelivery of tetra-substituted carboxyl aluminum phthalo- ranging from about 1.2 to 4.6. A chemotherapeutic drug DOX
cyanine (AlC4Pc) for PDT and small Pd nanosheets for was then noncovalently adsorbed on the mesoporous structure
PTT.140 While the PS was covalently conjugated to a of MSNs. The Ce6-doped mesoporous silica nanorods
mesoporous silica network, the Pd nanosheets were coated (CMSNR) with the highest aspect ratio was found to have
on the surface of mesoporous silica via coordination and highest Ce6 loading efficiency of 7.51% (w/w) as well as faster
electrostatic interaction. As both AlC4Pc and Pd nanosheets uptake in to the cancer cells. In vitro PDT using CMSNR
displayed maximum absorptions in the 600−800 nm region, the under 660 nm (5 mW cm−2 for 1 h), showed comparable or
fabricated 170 nm nanocomposites could be excited at the same slightly better cell killing effects compared to free Ce6.
time using a single 660 nm laser irradiation. Viability of HeLa Moreover, the synergistic effect of CMSNR/DOX was found
cell incubated with the dual functional nanocomposites (200 μg to be much stronger than the monotherapies both in vitro and
mL−1 for 12 h) dropped to below 35% following 10 min in vivo. The tumor growth inhibition in 4T1 tumor models
irradiation at power density of 0.5 W cm−2, which was following intratumoral injection of CMSNR/DOX and 660 nm
significantly higher than the means of either PTT or PDT, thus light irradiation (5 mW cm−2 for 1 h) was 60%, compared to
displaying a synergistic effect. 42%, 12%, and 27%, for Ce6+DOX (light), CMSNR (light),
Very recently, Yang et al. reported an interesting approach and CMSNR/DOX (light) groups, respectively.
for precisely modulating the morphology of MSNs by varying 5.1.2.2. Gold Nanoparticles. Gold nanoparticles (AuNPs)
the amount of organic PS molecules covalently doped in the possess a combination of unique properties such as the surface
silica matrix (Figure 10).141 In their study, the increase in the plasmon resonance (SPR) effect and the ability to convert light
R DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

energy to heat for PTT applications, due to which they have immune systems and were channeled into the excretory organs
gained much popularity in the development of combinatorial where it was finally removed from the body by renal and
hyperthermia-phototherapy agents. Moreover, due to localized hepatobiliary routes. As seen with other nanocarriers, develop-
surface plasmon resonances, field enhancement of the incident ment of multifunctional AuNP offering synergistic therapeutic
light around AuNPs could be used to increase the excitation approaches such a photothermal and PDT, has been the new
efficiency of the accompanying PS. Russell et al. was the first to trend in this field. The earlier reports however used two
report the synthesis of a three-component (PS/gold/phase different excitation wavelengths to accomplish PTT and PDT.
transfer reagent) system, where a phase transfer reagent For example, Jang et al. demonstrated a 95% reduction in
(tetraoctylammonium bromide; TOAB) was utilized to stabilize tumor growth in vivo using a gold nanorod AlPcS4 complex
phthalocyanines (Pc) on AuNPs.142 The developed 2−4 nm excited by 810 and 670 nm lasers.148 Khlebtsov et al. developed
construct generated 1O2 with enhanced quantum yields composite nanoparticles consisting of a gold−silver nanocage
compared to the free Pc, due to the presence of TOAB that core and a mesoporous silica shell functionalized with the PS
helps to increase the triplet energy transfer to molecular (Yb-2,4-dimethoxyhematoporphyrin) for in vivo PDT applica-
oxygen. In a follow-on work, they reported in vitro uptake and tions.149 The synthesized nanocomposites generated 1O2 under
photodynamic efficiency of these AuNPs in HeLa cells.143 The excitation at 630 nm and produced heat under laser irradiation
AuNP conjugates were effectively taken up by the cancer cells, at the plasmon resonance wavelength (750−800 nm). In order
and resulted in greater than twice the amount of cell-death to combine and simultaneously activate the two treatment
(57%) compared to free Pc (26%), which could probably be approach, Gao et al. developed a nanocomplex using a lipid-
attributed to the 50% enhancement of 1O2 quantum yield loaded HB to coat gold nanocages.150 Such an assembly,
observed for the Pc-AuNP. Subsequently, in vivo PDT efficacy combined with excitation using a 790 nm NIR two-photon
at 600−700 nm (157 J cm−2), following intravenous injection laser, resulted in one-off administration and excitation of both
of C11Pc (phthalocyanine derivative) conjugated AuNP was the gold nanocage, that convert light to heat energy and the
studied in amelanotic melanoma (B78H1 cells) subcutaneously accompanying PS, that generate ROS simultaneously. Similarly,
transplanted on mice.144 Compared to free C11Pc, AuNP- Seo et al. recently developed MB loaded mesoporous silica
C11Pc conjugates were found to target cancer tissues more coated gold nanorod for use in cancer imaging and PTT/PDT
selectively and to induce more extensive PDT response by dual therapy.151 The PS was physically adsorbed on the pores
promoting an antiangiogenic response by causing extensive of the silica shell. The excellent plasmonic properties of gold
damage to the blood capillaries and the endothelial cells. nanorods at NIR, enhanced the NIR light-induced SERS
However, the AuNP-C11Pc conjugates were taken up by liver performance of the embedded PS molecules by a factor of 3.0 ×
and spleen, with a prolonged persistence in the liver without 1010. Upon irradiation with NIR light at a single wavelength
any apparent decrease of the PS, for up to a week. Thus, (780 nm), the viability of CT-26 cells decreased to 31% for
suitable strategies to limit the accumulation of the nanoparticle- cells transfected with bare gold nanorods, while it dropped to
associated PS in important organs such as liver and spleen 11% for the MB loaded nanocomposites, clearly demonstrating
should be devised. As a subsequent step the same group the advantage of synergistic combination of the photothermal
reported targeted delivery of PEGylated AuNP-C11Pc and photosensitizing effects. It is to be noted that, unlike PDT
conjugates to breast cancer cells, by attaching anti-HER2 which is dependent on the availability of tissue oxygen, the
monoclonal antibodies to the PEG chain (HS-PEG-COOH PTT effect is oxygen-independent and hence the hypoxia
3000).145 In vitro experiments demonstrated selective targeting environment induced by PDT reaction does not affect PTT. In
of the 4-component “antibody-C11Pc-PEG-AuNPs” conjugate their excellent review, Liu et al. presented a very detailed and
to breast cancer cells that overexpress HER2 epidermal growth in-depth account of various nanoparticles used in PTT and
factor receptor and its efficacy in PDT applications, although PDT, as well as combination of PDT and PTT.152 Wang et al.
no in vivo results were presented. hypothesized that by adjusting irradiation time, an early phase
Cheng et al. evaluated covalent and noncovalent attachment PDT effect can be coordinated with a late-phase PTT effect to
of silicon phthalocyanine 4 (SiPc4) on PEGylated AuNP and achieve a better synergistic treatment efficacy.153 They
found that, in contrast to slow intracellular release and reduced synthesized a PEGylated gold nanorod, with Ce6 molecules
drug efficacy of covalently attached PS, noncovalent adsorption chemically conjugated to the PEG chains. The utility of the
showed efficient drug release into HeLa cancer cells by nanoconjugate was tested both in vitro and in vivo on MDA-
membrane-mediated diffusion.147 Subsequently, they explored MB-435 tumor bearing mice model. The tumor was exposed to
the drug delivery mechanism and pharmacokinetics following 671 nm laser (1.0 W cm−2) for 6 min, 4 h post intratumoral
intravenous administration of noncovalently bound PEG-SiPc4- administration of the nanoconjugates. A significant reduction in
AuNP conjugates over a period of 7 days.146 In vivo tumor volume was achieved post laser treatment with Ce6
experiments revealed enhanced accumulation of SiPc4 loaded PEGylated gold nanorods in comparison with free Ce6
molecules in tumor sites of cancer-bearing mice and dramatic or bare PEGylated gold nanorods, demonstrating significantly
decrease of the drug delivery time from 2 days to 2 h (Figure improved anticancer effect of the combinatorial approach.
11). Moreover, noncovalent attachment of PS to AuNP 5.1.2.3. Magnetic Nanoparticle. With further improvement
resulted in efficient release and penetration of the PEG- in the field of nanotechnology and nanomedicine, conceptu-
SiPc4-AuNP conjugate fast and deep into the tumors. Although alization and development of multifunctional theranostics
Au concentration in most tissues reached its maximum by 4 h probes became the next big wave in the field of PDT. Now,
followed by its gradual decrease over time, Au concentration in the success of PDT depends on choosing the apt time at which
liver and spleen continued to accumulate over time to reach a the light irradiation is to be performed when there is the highest
maximum at the latest time point measured, 7 days. concentration of the PS in the tumor compared to the normal
Biodistribution results suggested that the PEG-SiPc4-AuNP tissue. This could be realized by using noninvasive visualization
conjugates were removed from both the cardio−pulmonary and techniques that allow quantification of PS within the tumor.
S DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 12. (A) Schematic illustration for the synthesis and structure of IONC−PEG−Ce6. (B) SEM image of as-synthesized IONCs. Inset: TEM
image of IONCs. (C) T2-weighted MR images and T2 relaxation rates (R2) of IONC−PEG aqueous solutions at different Fe concentrations. (D)
UV/vis/NIR spectra of IONC−PEG, free Ce6, and IONC−PEG−Ce6 at different feeding Ce6 concentrations. (E) Fluorescence images of IONC−
PEG, IONC−PEG−Ce6, and Ce6 aqueous solutions at different excitation wavelengths (661 and 704 nm). The exposure time was 100 ms. (F)
Relative viabilities of 4T1 cells treated with various concentrations of IONC−PEG, Ce6, and IONC−PEG−Ce6 with light irradiation. (G) Tumor
growth curves of different groups of tumors after various treatments indicated. Error bars were based on SD of six tumors per group. MF: magnetic
field; L: light. (H) Representative photos of mice after various treatments. White and red arrows point to tumors without and with magnetic
targeting, respectively. Reproduced with permission from ref 159. 2011 American Chemical Society.

Magnetic resonance imaging (MRI) is one such promising phospholipids. The polymeric shell also serves as a suitable
noninvasive, harmless (no radiation exposure) visualization surface to further conjugate it with various biomolecules for
technique, in terms of its high spatial resolution and anatomical targeted delivery. In 2005, Kopelman et al. combined MRI
definition.154 In order to visualize the tumor and quantitate the contrast enhancement with targeted PDT, by constructing PAA
amount of PS uptake in the tumor, the PS was conjugated to a core nanoparticles encapsulating clinical PS, Photofrin, and iron
MRI contrast agent and encapsulated in a nanocarrier. Among oxide.155 The nanoconstruct was further surface functionalized
the broad spectrum of nanoscale materials available, iron oxides with PEG and integrin-targeting RGD peptide to enable
exhibiting superparamagnetism, have been most commonly simultaneous cancer targeting, detection, therapy, and monitor-
used in conjunction with PDT. Generally, the magnetic ing. A similar technique was adopted by Reddy et al. in which a
nanoparticles in the core are shielded from the surrounding vascular homing peptide, F3 was conjugated to a PAA core
environment with polymers like PEG, dextran, chitosan, PEI, or nanoparticle encapsulating Photofrin and iron-oxide/fluores-
T DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 13. Multifuntional theranosomes (A) Left: MRI scans of TC-1 tumors before and 48 h after PBS intratumoral injection (control group).
Right: MRI scans of tumors before and 48 h after theranosome intratumoral injection. (B) Normalized tumor growth curves in mice intratumorally
injected with theranosomes (25 μM m-THPC) or m-THPC (25 μM) followed by light exposure (λ = 630 nm, 30 J cm−2, 77 mW cm−2 for 390 s) 20
h later. All error bars reflect the SEM (n = 5). * and ** indicate p < 0.05 and p < 0.01, respectively, versus untreated control at day 7. (C) False-color
images of fluorescence emission from nude mice bearing TC-1 tumor before and after theranosome or m-THPC intratumoral injection, as a function
of time. Reproduced with permission from ref 163. 2011 American Chemical Society.

cent imaging agent.156 In both studies, the presence of targeting emerged as a new tumor-targeting strategy.160 In this technique,
agent enhanced the ability of the nanoconstruct to specifically systemically applied magnetic nanoparticles carrying therapeu-
target the tumors as revealed by MRI, thus increasing the tic agents would be attracted by the MF applied on the tumor,
efficiency of tumor killing post PDT. The ease of synthesis of resulting in enhanced enrichment of therapeutic agents in
silica nanoparticles with desired shape, size, and porosity, as targeted tumor region, consequently resulting in a significantly
well as its permeability to 1O2 has allowed incorporation of improved therapeutic effect compared to passive tumor
magnetite nanoparticles and PS into silica-based nanoparticles. targeting that is simply based on the EPR effect. Additionally,
Chen et al. described the synthesis of a multifunctional silica magnetic targeting may be applicable to a wide range of solid
nanoparticles (20−30 nm) encapsulating a porphyrin derivative tumors regardless of the expression of cancer linked receptors/
and iron oxide nanoparticles by microemulsion and sol−gel markers, unlike active tumor targeting strategies that is based
methods, with a PS encapsulation efficiency of 20.8%.157 The on the ligand−receptor binding. Along these lines, Sun et al.
nanoconstruct possessed good biocompatibility and 1O 2 synthesized porphyrin derivative encapsulated chitosan shell
generating ability, to cause remarkable photodynamic anti- surrounding an iron oxide nanoparticle via chemical copreci-
tumor effects. However, its ability as a MRI contrast agent was pitation method.161 The core−shell structured nanoparticles
not proven in vivo. Benachour et al. utilized active targeting of with a size range of 20.6 ± 5.1 nm and PS encapsulation
small silica-based nanoparticles with gadolinium oxide core for efficiency of ∼65.4%, were administered intravenously to
MRI and a chlorin based PS, as well as a peptide targeting xenograft mouse tumor model. The magnetic nanoparticles
neuropilin-1 (NRP-1) overexpressed in tumor vasculature.158 were targeted to the tumor by externally localized magnets and
In vivo studies on rats bearing an orthotopic human brain PDT was performed when a typical low intensity T2 weighted
tumor (U87) presented tumoritropic accumulation of the image was observed in tumor area, thereby achieving excellent
nanoparticles, following intravenous administration due to the tumor targeting, imaging and therapeutic efficiency in vivo.
presence of tumor vasculature targeting peptides, as revealed by Huang et al. prepared magnetic nanoparticles covalently
real-time MRI analysis. conjugated with Ce6 for simultaneous targeted PDT and in
Around this time, use of physical forces such as an external vivo dual-mode imaging (NIR fluorescence imaging and
magnetic field (MF) for enhancing the tumor localization MRI).162 The synthesized nanoparticles with a size range of
U DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

10 ± 2 nm and PS loading efficiency of ∼88.68%, had good


biocompatibility and remarkable photodynamic efficacy upon
irradiation. Furthermore, the nanoparticles were found to target
the tumor with an externally applied MF and their
biodistribution could be optically followed by NIR fluorescence
imaging. Similarly, Li et al. reported of 100 nm sized iron oxide
nanoclusters (IONC) exhibiting superparamagnetic property,
functionalized with PEG (Figure 12).159 Further loading of Ce6
was found to red shift the absorption peak of Ce6 at 650 to 700
nm. In vitro PDT at under 704 nm light irradiation (5 mW
cm−2 for 30 min, 9 J cm−2) showed remarkable concentration
dependent phototoxicity, owing to the enhanced uptake of
IONC-PEG-Ce6 compared to free Ce6. Additionally, enhanced
targeting of IONC-PEG-Ce6 post IV administration in a 4T1
mouse tumor model was achieved by subjecting the tumor to
MF, as confirmed by the reduction in the averaged T2 signal
intensity by 94.4% as compared to only 11% in the tumor that
was not subjected to MF. In vivo PDT of MF controlled tumor
at 704 nm (5 mW cm−2 for 1.5 h, 27 J cm−2) exhibited
significant tumor growth delay compared to the tumor that was
not subjected to MF, revealing highly effective tumor homing
ability of the multifunctional magnetic PDT agent.
Recently Silva et al. was the first to report proof of principle
of designing a natural, cell-derived, submicrometer vesicle as
multifunctional theranostic nanovector, named therano-
somes.163 Their strategy involved engineering of the parent
cells to enclose both iron oxide nanoparticles and a PS (m-
THPC), which were then stimulated to generate nanovesicles
(mean vesicle size of 332 ± 94 nm derived from transmission
electron microscopy (TEM)) laden with both agents conferring
magnetic and optical responsiveness, allowing therapeutic and
imaging functions. The MR scans following intratumoral
injection of theranosome at a density of 3 × 108 per μL
containing m-THPC concentration of 25 μM and iron
concentration of 5 mM exhibited large zone of hypointense
signal within the tumor, confirming its intratumoral loacaliza-
tion (Figure 13). At the same time, fluorescence imaging Figure 14. Schematic illustration of photodynamic reaction upon light
revealed an increase in fluorescence signal 20 h after injection, irradiation on (A) fullerene and (B) TiO2 nanoparticle. (•OH,
for both theranosomes and free m-THPC, perhaps due to m- hydroxyl radical, O2•−, superoxide anion, HO2•−, hydroperoxyl radical
and H2O2, hydrogen peroxide).
THPC diffusion toward the tumor surface. However, signals
from theranosomes were restricted to the tumor site while
extratumoral extravasation was observed for the free drug. In their extended π-conjugation, fullerenes absorb light in the
vivo PDT following intratumoral administration of therano- ultraviolet (UV) or blue regions of electromagnetic spectrum to
somes in TC-1 tumor bearing mice revealed significant form long-lived triplet state and generate ROS upon
reduction in tumor growth compared to nontreated control illumination, allowing them to act as PSs. In the recent years
at day 7. However, systemic administration of theranosomes its potential as PS has been widely studied due to several
needs to be addressed to gain insight in the translational advantages. Fullerenes are comparatively more photostable and
potential of this approach. not easily susceptible to photobleaching.164 They undergo both
Type I and Type II reactions generating both free radicals and
5.2. Nanoparticles as Downconverting Photosensitizers 1
O2. Furthermore, they do not breakdown and are excreted
Certain nanoscale materials have the ability to generate ROS, intact. However, the chief disadvantage of fullerenes is their
due its unique optical absorption properties, thus allowing them optical absorption properties; as they absorb light where the
to behave as PSs by itself. In the following section, the tissue penetration depth of light is shortest due to a
properties and application of three such nanomaterials are combination of light absorption by endogenous chromophores
discussed. and light scattering by cellular structures. Second, they are
5.2.1. Fullerenes. Buckminsterfullerene (or bucky-ball) is a innately insoluble in water, due to which they severely
carbon allotrope discovered in 1985, with a spherical shape and aggregate in aqueous conditions, hampering its biological
usually composed of 60 (C60) or 70 (C70) carbon atoms application. This shortcoming of the fullerenes have been
(Figure 14A). It has a hollow cage-like fused structure with sp2 overcome by surface modification or supramolecular ap-
carbon atoms arranged in hexagons and pentagons. The proaches such as PEGylation,165 encapsulating in liposomes,166
diameter of the fullerene cage is approximately 7−10 Å, thus micelles,167 or chitosan.168
making it less likely to be taken up by the macrophage cells As with other nanoparticles used in PS delivery, fullerenes
compared to other larger conventional nanoparticles. Due to have also been modified to carry imaging agents to bestow
V DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

multifunctionality. Liu and others developed a theranostic photokilling of malignant HeLa cells grown on a polarized
hybrid system in which diethylenetriaminepentaacetic acid illuminated TiO2 film electrode.177 Although the initial
(DTPA) was attached to the terminal group of PEGylated C60 experiments showed successful photocatalytic cell killing, the
(C60-PEG-DTPA) and subsequently mixed with gadolinium use of a TiO2 electrode system was not considered to be
acetate solution to obtain Gd3+-chelated C60−PEG−DTPA− feasible in vivo. Hence, a series of in vitro studies on HeLa cells
Gd.165c Upon light irradiation (400−500 nm, 53.5 J cm−2) followed using colloidal TiO2 particles in suspensions, as it
following intravenous administration of C60−PEG−DTPA− could be incorporated by the living cells and had extremely
Gd into tumor-bearing mice, there was significant antitumor large surface area, thus resulting in higher photocatalytic
PDT effect, which correlated with tumor accumulation as reaction rates.172c,178 Subsequently, the detailed mechanism of
detected by the enhanced intensity of MRI signal. Recently, UV induced phototoxic effect of TiO2 nanoparticles was
functionalized fullerenes targeted to cancer cells have been reported on a series of human cancer cells such as bladder
developed and its potential in PDT is being investigated. In cancer cells (T24),179 monocytic leukemia cells (U937),180
2013, Liu et al. developed trimalonic acid-modified C70 adenocarcinoma cells (SPC-A1),181 colon carcinoma cells (Ls-
fullerenes (TF70) conjugated with aptamer R13, specifically 174-t),182 breast epithelial cancer cells (MCF-7, MDA-MB-
targeting epithelial growth factor receptors (EGFR) overex- 468),176b and glioma cells (U87).183
pressed in certain cancer cells.169 Incubation of R13 conjugated Only a few studies have reported the in vivo application of
TF70 with A549 lung cancer cells, displayed good binding TiO2 nanoparticles for PDT in animal models. Generally, the
ability to the EGFR expressing A549 cells. Further studies also TiO2 particles were either administered intratumorally or
revealed preferential localization of the fullerene conjugate in subcutaneously over the tumor tissue, rather than intravenous
the lysosome and facilitated efficient cell-kill under illumination delivery due to its insolubility and aggregation issues in
with white light. In a yet another approach, malonic acid C60 physiological environment, which means that it could be easily
derivative (DMA-C60) modified with DSPE-PEG2000-malei- recognized and removed from the circulation by the cells of the
mide was conjugated to Asn-Gly-Arg (NGR) peptide, that MPS and ultimately reduce its accumulation in the tumor.
selectively targets CD13 isoform overexpressed in tumor Following administration of TiO2 nanoparticles, the tumors
vasculature and certain tumor cells.170 2-Methoxyestradiol were irradiated with UV light after the skin was surgically
(2ME), a nonpolar endogenous metabolite of 17β-estradiol removed to expose the tumor. This is because the penetration
exhibiting antiproliferative and antiangiogenic activities, was depth of UV light (300−400 nm) into the tissue is limited to a
then attached to DMA-C60-NGR through physisorption, to few fractions of a millimeter.184 Cai et al. reported significant
form a novel tumor-targeting drug delivery system DMA-C60− suppression of HeLa cells implanted in nude mice for up to 30
2ME-NGR for synergistic enhancement of cancer therapy. The days, compared with those receiving TiO2 alone or UV
presence of targeting peptide enhanced cell uptake of DMA- irradiation alone.172c Furthermore, when tumors received a
C60−2ME-NGR that localized in the cytoplasm and also second repeat treatment of TiO2 and UV light irradiation on
promoted a faster 2ME transfer into cells, exerting a stronger day 13, the tumor growth was inhibited more dramatically.
inhibition on MCF-7 breast cancer cells. The results revealed a Similarly, Kubota et al. also reported a drastic delay in T-24
synergistic enhancement of breast cancer therapy using PDT bladder cancer tumor implanted in nude mice up to 30 days
induced by DMA-C60-NGR and 2ME. post-PDT using TiO2 particles under UV light illumination.179
5.2.2. Titanium Dioxide Nanoparticle. Titanium dioxide Likewise, Wang et al. found that the combination of TiO2 and
(TiO2), also known as titania, is the naturally occurring oxide of UV irradiation was more effective not only in reducing tumor
titanium. Ever since the discovery of photoinduced decom- growth but also in promoting survival.183 However, it should be
position of water on TiO2 electrodes under UV light was noted that this technique was not effective in retarding a tumor
made,171 it has found multifunctional applications which that had grown beyond a certain size.
underpin many important energy-conversion processes, includ- 5.2.3. Zinc Oxide Nanoparticle. Nanosized zinc oxide
ing photocatalysis, photodegradation, environmental remedia- (ZnO), a well-known photocatalyst comparable to TiO2, is
tion, water splitting for hydrogen fuel, CO2 reduction, self- frequently found in industrial products including cosmetics,
cleaning coatings, electrochromic devices, sensors, and low-cost paints, and medical materials. ZnO has almost the same band
solar cells. gap energy (3.2 eV) as TiO2 and has a similar photocatalytic
In recent years, TiO2 has been regarded as potential activity. Li et al. compared the phototoxic effect of ZnO
photosensitizing agent for PDT due to its low toxicity, nanoparticles of three different sizes (20, 60, and 100 nm)
physiological inertness, excellent biocompatibility, and unique following irradiation with the UVC (254 nm; 0.1 mW cm−2 for
photocatalytic activity.172 Under irradiation with UV light with 180 s) and studied the synergetic cytotoxicity of the anticancer
energy greater than the band gap energy of TiO2 (i.e., shorter agent daunorubicin (DNR) with ZnO nanoparticles against
than 385 and 400 nm corresponding to a band gap energy of hepatocellular carcinoma cells (SMMC-7721) in vitro.185 The
3.23 and 3.06 eV for anatase and rutile polymorph of TiO2 phototoxic effect was found to be size dependent. Meanwhile,
respectively), the electrons in the valence band of TiO2 are significantly greater cell-kill was achieved when SMMC-7721
excited to the conduction band, thus resulting in the formation cancer cells were treated with a combination of ZnO
of photoinduced hole−electron pairs (Figure 14B). These nanoparticles and daunorubicin. In 2010, Hackenberg and co-
photoinduced electrons and holes possess strong reduction and workers studied the photocatalytic effects of ZnO nanoparticles
oxidation properties that can interact with surrounding O2 and on human head and neck squamous cell carcinoma (HNSCC)
H2O molecules to generate various ROS, such as hydroxyl cell lines and nonmalignant cells in vitro.186 Studies revealed
radical (OH•),173 superoxide anion radical (O2•−)174 and that ZnO nanoparticles at concentrations of 0.2 and 2 μg mL−1
hydrogen peroxide (H2O2),175 which react with biological in the presence of UVA (20 mW cm−2 for 15 min) presented
molecules, such as lipids, proteins, and DNA, and eventually kill the optimal conditions for cancer cell-kill, mainly via a
cancer cells.176 Fujishima et al. was the first to report combination of late apoptosis and necrosis. In addition to its
W DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

photosensitizing capabilities, ZnO was also used as a delivery


agent of anticancer drugs for combination therapy. Hariharan et
al. fabricated PEGylated ZnO (ZnO/PEG) nanospheres loaded
with anticancer drug of DOX and studied both its antibacterial
and antitumor activity.187 The DOX loading and encapsulation
efficiency on ZnO/PEG nanospheres were found to be 70.35 ±
6.20% and 21.34 ± 2.4%, showing its potential as drug delivery
agents. DOX-ZnO/PEG nanocomposites proved to be capable
of photodynamically inactivating of Gram-positive micro-
organisms under visible light. Furthermore, the nanocomposites
exhibited dose-dependent toxicity toward HeLa cell lines, as
well as demonstrated improved therapeutic efficacy upon UV
irradiation, thus minimizing the side-effects.
5.3. Nanoparticles as Energy Transducers
Certain nanoparticles are not just carriers of PS but they also
actively participate in energy transfer to the attached PS.
Generally, the energy harnessed from light is then transferred
to the PS associated with it, thereby offering the possibility of
indirectly activating the PS using light at wavelengths that it
normally does not absorb.
5.3.1. X-ray Activatable Nanoparticle. The first report
on X-ray activatable nanoparticles was back in 2006 by Chen et
al., where they described a new approach to treat cancer
through a combination of radiation therapy and PDT, termed
as self-lighting PDT.188 The rationale behind this technique was
to utilize the stronger absorption of the porphyrins at around
400 nm (Soret band) rather than the weak absorption at
around 600−800 nm (Q-band). This is because excitation of
the PS at the Soret band can be much more efficient in the
generation of ROS. Unfortunately, attempting to activate PS
through absorption at the Soret band will compromise the
Figure 15. Schematic illustration of energy transfer from X-ray
clinical usefulness of PDT, as UV/blue light has minimal activatable scintillation nanoparticle to the attached PS.
penetration in tissues. Hence, red light (>600 nm) is often
utilized to activate the weak Q-band of the PS. In an effort to
utilize the strongly absorbing Soret band of PS at the UV/blue emitting strong luminescence.192 Again, nanoparticles with
region, scintillation or persistent luminescence (“after glow”) small diameters are preferred as they have a weak electron−
nanoparticles (e.g., BaFBr:Eu 2+ , Mn 2+ , LaF 3 :Ce 3+ , and phonon coupling and consequently exhibit greater lumines-
LaF3:Tb3+), with attached porphyrin PS were used as in vitro cence efficiency due to a smaller Stokes shift.193 And last, in
PDT agent. These nanoparticles upon exposure to X-rays emit order to have efficient energy transfer, the distance between the
luminescence in the visible region (peaking at 400, 500, and donor and the acceptor should be less than 10 nm.
650 nm) of the spectrum activating the attached PS at both the The highlight of this approach is that as there is no limitation
Soret band and Q-band, resulting in the efficient generation of to the penetration depth achievable by X-rays in tissue,
1
O2 (Figure 15).188,189 In a pilot study, the energy transfer theoretically any deep-seated or solid tumors can be treated
following X-ray irradiation of water-soluble LaF3:Tb3+ nano- effectively by photoactivation of the attached PS by the X-ray
particles to the attached meso-tetra(4-carboxyphenyl)porphyrin activatable nanoparticles. Moreover, the synergistic effect of low
(mTCP) was demonstrated by fluorescence quenching dose radiation therapy and PDT is expected to enhance the
techniques.190 In a very recent mechanistic study by the same cancer cell killing effect. Lastly, as the nanoparticles are self-
group, Ce3+-doped lanthanum(III) fluoride (LaF3:Ce3+) nano- illuminating especially if it emits an “after glow”, there is no
particles with strong green emission at approximately 520 nm need of an external light source, making this therapy highly site
upon excitation with X-rays, were encapsulated in poly(D,L- specific. Afterglow is the persistent luminescence that remains
lactide-co-glycolide) microspheres along with PpIX.191 Upon X- even after the X-ray irradiation is turned off. Generally, an
rays irradiation (90 kV), energy transfer from afterglow nanoparticle also has scintillation luminescence, but a
LaF3:Ce3+nanoparticles to PPIX resulted in 1O2 generation scintillation nanoparticle might not necessarily emit an
and cancer cell kill, due to oxidative stress, mitochondrial afterglow. Use of nanoparticles with such an “afterglow” and
damage and DNA fragmentation. Thus, the energy transfer long decay lifetimes could greatly reduce the amount of
from the X-ray scintillation nanoparticles to the PS is very radiation dose required for initial excitation of the scintillation
critical and this requires very good overlap between the nanoparticle and prolong the PS excitation period. However, in
emission peaks of the nanoparticles and the absorption or the earlier studies X-rays with energy in the kilovoltage range
excitation peaks of the PS. Second, such nanoparticles must were used, while beams with energy in the megavoltage range
have strong luminescence efficiency. Generally, higher atomic are normally used in the clinics for conventional radiotherapy.
number phosphors usually have greater radiation stopping It was not reported whether the scintillation nanoparticles
power and higher absorption coefficient for radiation, thus could be excited by megavolatage (MV) X-rays, and if then,
X DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 16. Schematic illustration of energy transfer from nanoparticles to PS and corresponding simplified energy diagrams. (A) QD transferring
energy to PS via FRET, (B) gold nanorod as a two-photon absorbing nanoparticle.

there will be enough energy transfer to the conjugated PS for Although the QD by itself did not generate 1O2 and the
significant cell killing. To address these critical questions, Yang quantum yield of 1O2 both via direct excitation of PS or
et al. developed a amine-functionalized PEGylated quantum dot indirectly through the FRET mechanism was relatively low, the
(QD-CdSe core with ZnS shell) conjugated with porphyrin and advantage of this technique was that several PS molecules could
studied the energy transfer and cell killing effect following be coupled to QD to improve the FRET efficiency. Recently,
irradiation with 6-MV X-rays.194 A significant cell killing was Hsu and co-workers demonstrated that self-illuminating QDs
observed with combined radiation and PDT treatment can serve as a versatile light source for activation of PS and may
compared to radiation alone. The linear relationship between overcome the limitation of light penetration for PDT.198 They
the radiation dose rate and QD excitation allows prediction of fabricated a Renilla luciferase-immobilized QDs (QD-RLuc8)
light dosimetry, which would be useful in the translation of the conjugate. After the addition of coelenterazine (the substrate of
novel conjugates for clinical application. RLuc8), energy is released from RLuc8 and transferred to QDs
5.3.2. Quantum Dots. Semiconductor QDs holds great via bioluminescence resonance energy transfer (BRET), leading
promise in PDT applications. Depending on the size (ranging to self-illumination of QD-RLuc8 conjugate at 655 nm. This
from 1 to 6 nm) and composition, the QDs can have unique bioluminescent photon emitted from QD-RLuc8 conjugate, in
optical and emission properties that can be precisely tuned turn excites the Foscan-loaded micelles and successfully
from the UV to the infrared region. The surface of QDs can be generated ROS achieving ∼50% cell kill in vitro and delayed
modified and functionalized to make them both water-soluble tumor growth in vivo.
and biocompatible. Furthermore, they possess a large transition 5.3.3. Two-Photon Absorbing Nanoparticles. Certain
dipole moment, making them strong light absorbers, ideal for molecules with two photon absorption (TPA) cross-section can
PDT applications. QDs could act as energy donors and transfer simultaneously absorb two low-energy photons and emit higher
the energy to cell molecules like triplet oxygen, reducing energy photons (Figure 16B). Such dyes offer new perspectives
equivalents or pigments, potentially inducing generation of for the treatment of solid tumors using PDT, as they can be
ROS leading to cell kill. Although Samia et al. reported that excited in the NIR region allowing deeper light penetration in
cadmium selenide (CdSe) QDs could be a potential PS on its tissues. Two-photon PDT (TP-PDT) allows 3-dimensional
own and generate 1O2, the quantum yield of CdSe-generated selectivity to precisely target tumor cells, thus preventing
1
O2 was rather low (5% versus 40−60% by conventional damage to adjacent healthy tissues. Conventional PSs usually
PSs).195 However, the authors also reported that a more have low TPA cross sections in the biological spectral window
efficient (77%) energy transfer via FRET mechanism occurs (i.e., 700−1000 nm), requiring high excitation powers that are
between a QD and PS (Pc4) when excited at 488 nm, due to close to the threshold of tissue photodamage. Thus, even
the close steric proximity between the two structures (Figure though the activation of some PSs such as ICG falls within the
16A). Here, Pc4 was excited by CdSe-QD emission at 568 nm NIR range (∼800 nm),199 their quantum yield of 1O2 remains
and thus enabling the use of an excitation wavelength where the low due to insufficient triplet formation.200 Thus, most of the
PS alone does not absorb in a normal scenario. However, initial studies combined TPA dyes with PS to convert the
insolubility of the conjugate in water hampered its application energy emitted from TPA dyes to activate PS for PDT.
in biological systems. Later Shi et al. fabricated water-soluble However, dark-toxicity of these dyes was often a major concern.
QDs with porphyrin PS, meso-tetra (4-sulfunatophenyl) As a solution to this problem, Kopelman and co-workers
porphyrin (TSPP) electrostatically attached to the QD.196 entrapped a TP dye 5,10,15,20-tetrakis(1-methyl 4-pyridinio)
Upon excitation at 355 nm, at which the PS will otherwise not porphyrin tetra(p-toluenesulfonate) (TMPyP) in PAA-based
absorb, the QD-PS hybrid nanocomposite was found to nanoparticles.201 A different approach was adopted by Prasad
generate 1O2. However, potential toxicity of synthesized and co-workers, where they coencapsulated a TP dye, 9,10-
uncapped QDs was the major drawback. Tsay et al. coupled bis[4′-(4″-aminostyryl)styryl]anthracene (BDSA) and HPPH
two different PS, Rose Bengal (RB) and Ce6 to a peptide, in a ORMOSIL nanoparticles.202 Here, the BDSA acted as a
which was in turn attached to CdSe/CdS/ZnS QDs.197 two-photon absorbing dye unit (energy donor) that indirectly
Y DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 17. Schematic of energy transfer from UCN phosphor to the attached PS and corresponding energy diagram.

excited HPPH (energy acceptor), via FRET and cytotoxic effect effectively excited via TP irradiation at 1030 nm and FRET
under two-photon irradiation (850 nm) was demonstrated. efficiency of up to 82% was obtained when multiple Ce6
Velusamy et al. synthesized quadrupolar type TPA chromo- molecules were bound to QD. However, none of these reports
phores exhibiting remarkably large TPA cross section values of backed up their results with in vitro or in vivo PDT studies.
about 7000 Goeppert−Mayer units (GM) at 800 nm in Conjugated polymers have large delocalized π-conjugated
toluene.203 To further improve its biocompatibility and backbones and are known to have large TPA coefficients, high
solubility in aqueous solution, the compound was encapsulated fluorescence quantum, and good biocompatibility, and nano-
in silica nanoparticles. In vitro PDT experiments at 800 nm particles made of such conjugated polymers were found to be
NIR irradiation for 3 min at 468 J cm−2 using macrophage cells promising materials for TP-PDT applications. Xu et al.
(Raw 264.7) killed about 40% of cells at a concentration of 100 encapsulated hydrophobic tetraphenylporphyrin (TPP) in
μg mL−1. Gary-Bobo and co-workers demonstrated efficacy of poly[9,9-dibromohexylfluorene-2,7-ylenethylene-alt-1,4-(2,5-
TP-absorbing porphyrin derivative covalently conjugated in dimethoxy)phenylene] (PFEMO) nanoparticles (∼40 repeat
mannose functionalized biocompatible MSNs in an in vivo units).209 Here, PFEMO nanoparticles (∼50 nm) served as
model.204 TPA properties of the PS were retained in the MSNs, both the hosting material and two-photon light harvesting
with giant TPA cross sections (up to 8 MGM) for a single complex and the TPA cross section of PFEMO was measured
MSN and induced significantly more cell death in vitro to be 2160 GM per molecule (54 GM per repeat unit), while
following irradiation at 760 nm by three scans of 1s duration the TPA cross section of TPP was 12 GM. The two-photon
each delivering a power of 80 mW. Intravenous administration emission of TPP was found to be significantly enhanced by up
of 16 mg kg−1 mannose-functionalized porphyrin loaded MSNs to 21-fold in the nanoparticles, and there was a significant
in HCT-116 xenograft mouse model, revealed that a single enhancement of 1O2 generation with 1% TPP-doped PFEMO
injection was sufficient to target these nanoparticles to the nanoparticles under TP excitation at 800 nm. This confirmed
tumor area, while TP-PDT treatment at 760 nm induced a energy transfer from PFEMO to TPP, thus demonstrating its
major reduction of the tumor size. Thus, the initial studies potential as photosensitizing agents for two photon-PDT and
utilized nanoparticles as a carrier of TP absorbing dyes/PS, related applications. Although the PFEMO nanoparticles
rather than actively participating in TPA. Thereafter, nano- displayed good stability in water, they aggregated in high
particles having large TPA cross sections, were utilized for TP ionic strength medium like PBS. To circumvent this issue of
excitation in a donor−acceptor system for indirect excitation of nanoparticle aggregation, the same group incorporated a
PS and one such example is QD-PS conjugate. Dayal et al. nonionic surfactant, polyoxyethylene nonylphenylether (CO-
conjugated Pc4 with CdSe QDs and demonstrated successful 520), in the polymeric nanoparticle, to confer hydrophilic
energy transfer to PS following two-photon excitation of the polyoxyethylene chains on the nanoparticle surface.210 In vitro
QDs at 1100 nm.205 Wen et al. fabricated water-soluble CdTe studies revealed negligible dark-toxicity in HepG2 cells.
QD as an energy donor to indirectly excite electrostatically However, under 800 nm laser excitation (630 J cm−2), the
attached mesotetra-4-sulfonatophenyl-porphine dihydrochlor- cell viability decreased to about 58%, demonstrating for the first
ide (TSPP), with an energy transfer efficiency of 82%, time the application of TP excitable PS-doped conjugated
demonstrating its potential in TP-PDT.206 Qi et al. covalently polymer nanoparticles for cancer cell killing in vitro.
attached water-soluble porphyrin on the biocompatible Gold nanorods are notable for their large two-photon
amphiphilic polymeric micelles encapsulating CdSe QDs, to absorption cross-section and tunable longitudinal plasmon
enable FRET to porphyrin upon two-photon excitation of QD band (Figure 15B). Very recently, Zhao et al. developed
at 800 nm.207 However, covalent conjugation of QD−PS composite nanoparticles by covalently binding porphyrin
conjugates required additional chemical procedures which are molecules (T790) to silica shell coated gold nanorods.211
not always feasible, while electrostatic interaction might be less The silica shell acted as a spacer to control the distance
stable in biological media. Recently, Skripka et al. exploited the between gold nanorod and the optimum two-photon excitation
hydrophobic interaction to design QD−PS complex, consisting fluorescence enhancement of these core−shell nanoparticles
of QD coated with PEGylated phospholipid layer which serves was achieved with a shell thickness 20 nm, by a factor of 11.8.
as a binding sites for nonpolar moiety of Ce6.208 The PS was 1
O2 generation capability and PDT effect of these core−shell
Z DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

nanoparticles under two-photon excitation was evaluated in UCN-PS system for PDT as well as methods utilized to
HepG2 cells and showed a 50% reduction in cancer cells with 8 incorporate multiple functionalities to the construct, in order to
min irradiation using femtosecond laser at 800 nm. Despite develop it into a multifunctional theranostic probe for
being a highly spatially focused technique, the requirement of simultaneous imaging and therapy for a “see and treat”
high excitation intensities and simultaneous absorption of two approach.
coherent NIR photons, reduces the probability and, thus, 5.3.4.1. Selection of UCNs and Photosensitizers. The UCN
efficiency of this approach. Furthermore, it excites a tiny tissue constructs used in PDT generally consists of three components
volume usually resctricted to a few femtolitres,212 often namely host, dopants, and PS. In the following section we will
resulting in the need of prolonged treatment time, thus limiting review the selection criteria of these three components, to
its clinical use especially in the treatment of large tumors. This generate an efficient UCN based PDT system.
underscores the need for a “nano-transducer” that can convert Host. The host material provides a crystalline lattice in which
deep penetrating low-energy NIR light to a higher-energy light the dopant ions are accommodated. Hence, there should be a
that matches the absorption spectrum of PS associated with it. close match between their lattices, to achieve high doping
In the following and final section of this review, we will discuss levels. Inorganic materials such as trivalent rare earth ions (La3+,
in detail the concept and principle behind developing UCNs Gd3+, and Y3+), alkaline earth ions (Ca2+, Ba2+, and Sr2+), or
that has the ability to harness energy from the NIR light and transition metals (Ti4+ and Zr4+) can be used as host
convert it to higher energy−visible or UV light, which can then materials.216 The optical properties of the UCNs such as
excite the PS associated within its close proximity. Although emission profile and upconversion efficiency depends on the
UCNs have many applications in biomedicine like bioimaging, host, and hence selection of host material is critical for efficient
biosensing, and drug or gene delivery, this article essentially PDT. In order to reduce energy losses during upconversion via
focuses on reviewing its application in the field of PDT. We will multiphonon relaxation and to maximize radiative emission, a
then present an overview of the recent reports, summarize its low phonon energy lattice is essential.217 The crystal structure
current limitations, and further discuss ways to augment PDT of the host material also influences the crystal field around the
efficacy in order to take this technology ahead from bench to dopant ions, resulting in different optical properties of the
bed-side. UCNs.218 The densely packed hexagonal (β-phase) materials
5.3.4. Upconversion Nanoparticles. UCNs are nano- allows efficient energy transfer between the dopant ions, thus
meter-sized materials that convert low energy light to high exhibiting higher upconversion efficiencies than their loosely
energy light through sequential excitation with multiple packed cubic (α-phase) counterparts.219 Thus, Jin et al. induced
photons, via an anti-Stokes emission process. Materials a cubic to hexagonal phase transition of the NaYbF4 crystal by
exhibiting such photon upconversion are usually comprised of Gd3+ ion (24%) doping, thereby enhancing the emitted blue
host lattices of ceramic materials, embedded with transition UCL by 30%.220 Another important factor is the phase-purity
metal, actinide or lanthanide ions like Yb3+, Er3+, and Tm3+.213 of the host where phase-pure hexagonal host materials with an
Such materials possess multiple “real” metastable electronic even symmetry exert uniform crystal-field around its dopant
states which can principally be used to generate upconversion ions and minimize the energy loss arising from crystal defects,
luminescence (UCL; Figure 17).214 In general, the lowest resulting in enhanced upconversion efficiency.219a Furthermore,
metastable state acts as an energy store and absorbs in the NIR the cationic size of the host also influences the upconversion
region to promote electrons to the first metastable state S1.214 efficiency such that a host with a smaller unit-cell volume has
A subsequent absorption of a second NIR photon promotes the greater crystal-field strength around its dopant ions and hence a
electrons from metastable state S1 to a much higher metastable higher upconversion efficiency.216 Despite the vast knowledge
state S3. Although, some internal conversion and relaxation of available on various host materials, NaYF4 has been the host
electrons back from S3 to S2 is possible, the electrons return to material of choice for UCN constructs in PDT. Perhaps, this
the ground state by emitting a higher energy photon. Due to could be due to the low phonon energies (∼360 cm−1),221
the uniqueness of this material it has found numerous biological smaller size and high chemical stability of this fluoride based
applications by virtue of its tunable emission, large anti-Stokes host material. A few studies have also reported the use of other
shift, sharp emission bandwidth, and high photostability.215 host materials containing Gd222 (NaGdF4) and Yb ions220
Recently, it has become increasingly necessary to develop new (NaYbF4) that can also function as MR and CT contrast agents
PSs that could be excited at NIR range, to augment the scope respectively, thus imparting additional imaging capabilities to
and applicability of PDT especially for the treatment of solid the UCN construct. Recently, Wang et al. synthesized
and large tumors. This is where the UCN technology becomes tetragonal-phase LiYF4 hosts with good crystallinity and high
very useful, because UCNs could act both as delivery agent of upconversion quantum yield and further demonstrated an
the PS as well as convert the NIR light to visible light, required improved upconversion efficiency with the increase in size, due
for the activation of associated PS at much deeper levels in the to reduced surface quenching effects of the UCNs.222c To
tissues. While, two-photon excitation requires simultaneous minimize the surface defects and resultant luminescence
absorption of two photons using a virtual state with a lifetime quenching effects, the UCN core may further be coated with
scale of femtosecond, upconversion process requires sequential an undoped matrix forming a shell such as NaGdF4220,223 or
absorption using a real metastable state with a lifetime in the CaF2.222b
order of microseconds. Again, the typical power densities for Dopants. The dopant ion is the key player in the process of
upconversion is usually in the range of 1−103 W cm−2 upconversion that actually performs the absorption and
achievable by continuous wave lasers, where as that of two- emission of photons. The emitted UCL largely depends on
photon absorption is 106−109 W cm−2. Hence, the efficiency of the type and the amount of ions doped. Rare earth metals, such
upconversion process is several orders of magnitude higher as lanthanides, are suitable candidates as dopants, as most of
than that of two-photon emission. In the following sections, we them (except La, Ce, Yb, and Lu) have appropriately spaced
will discuss in detail the different strategies adopted to design a multiple ladder-like metastable states, necessary for the
AA DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 18. Schematic illustration of various PS loading strategies in UCNs.

phenomenon of upconversion.214,217b This class of materials a system can be utilized for efficient energy transfer when PS
have 4f electrons spatially buried by lower energy 5s and 5p with a broad absorption bands such a fullerenes are used.
electrons resulting in a weak electron−phonon coupling.224 Photosensitizers. Zhang et al. was the first to report the use
Thus, multiphonon relaxation becomes less efficient due to of NaYF4:Yb3+,Er3+ UCNs with a thin layer of silica doped with
sufficiently large gap to the next lower energy level and thereby a lipophilic PS merocyanine-540 (MC-540), for PDT.230 Since
resulting in long-lived excited states (up to 0.1 s).214 The then many PSs, mostly hydrophobic (zinc phthalocyanine
UCNs used in PDT usually consists of at least two dopant ions (ZnPC), Ce6, HP, TPP, hypocrellin A (HA), Ppa, silicon
namely, sensitizer and activator. Such a codoped system enables phthalocyanine dihydroxide (SPCD) etc.) have been used in
us to enhance the upconversion efficiency. A commonly used combination with UCNs for PDT. A few water-soluble PS like
sensitizer ion is Yb3+ that has a relatively large absorption cross- MB and RB have also been chosen to be used in conjunction
section and a single metastable state.225 Furthermore, it has a with UCN technology. The selection of PS to be used in
reduced tendency of concentration-dependent quenching, combination with a particular UCN type is critical and there
hence increasing the concentration of Yb3+ could increase the must be a very close match between the UCN emission and
excitation probability of the commonly used activator ions like absorption maxima of the PS for efficient upconversion to
Er3+/Tm3+ via an energy transfer process. This efficient energy occur. Another important factor to be considered is the method
of PS loading and the proximity of PS molecule to the UCN
transfer occurs because the energy levels of Yb3+ and Er3+ are
core. As PS absorbs the upconverted light emitted by the UCN
perfectly in sync that they do not undergo phonon relaxation.
core, it has to be placed in close proximity to the UCN core for
Nevertheless, to minimize the loss of the excitation energy by
efficient generation of ROS for PDT. PS can be entrapped,
cross-relaxation process, the doping level of the activator is kept noncovalently adsorbed, and chemically or electrostatically
below 2 mol %, whereas that of the sensitizer is around 20 mol attached on the UCN surface. The different strategies adopted
% in such a codoped system.226 Such Yb−Er based UCNs to attach PS to UCN are discussed in detail elsewhere in this
usually give a bright green emission (around 550 nm) and a review.
weaker red emission (around 660 nm), the latter which is often 5.3.4.2. Surface Modification of UCN and Attachment of
utilized to activate the attached PS. In order to obtain a single Photosensitizer. Following the synthesis of hydrophobic UCNs
band, dark red emission for a higher FRET efficiency to red that usually involves preparation in organic solvents, it is
light absorbing PS, Mn2+ ions could be doped into the host transferred to aqueous phase before further utilizing it for
matrix.227 Another strategy involved increasing the Yb3+ ion biological applications. This is often done by surface coating or
concentration from the usual 20% to 25%. This was shown to modifying the core to impart hydrophilicity by addition of
enhance the emission intensity of red UCL band at 660 nm to stabilizers. The surface chemistry of UCN ultimately
about 2.69 folds higher than the green UCL band.228 Strong determines its solubility, stability, biocompatibility, pharmaco-
multicolour UCL could be obtained by separately doping kinetic properties, and capability of anchoring different
NaYF4 core and shell with Er3+ and Tm3+ (NaYF4:Yb3+, Er3+/ functional groups, targeting ligands as well as PS or other
NaYF4:Yb3+,Tm3+) instead of homogeneous codoping.229 Such drugs. To date the UCN surface chemistry has been improved
AB DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

by either polyethylenimine (PEI),231 PEGylation (addition of silica coating first described by Zhang et al.230 lacked this
PEG),232 surface coating with amphiphilic PEG block permeability, which could have resulted in decreased PDT
copolymers,232a,233 PEG-phospho-lipid,223 chitosan,234 BSA or efficiency. Second, surface modification by silica coating often
coating with silica.222a,b,226b,230,235 leads to particle agglomeration in physiological conditions.
PEI coating has the advantage of being a one-step “one pot” Mesoporous silica coating of UCN was found to have superior
hydrothermal process, whereby amine functionalized UCNs are properties than a simple amorphous silica coating, such as
synthesized that can be utilized for covalent conjugation of better solubility and greater efficiency of energy transfer, due to
other ligands via 1-ethyl-3(3-(dimethylamino)propyl)- its porous nature.235 Furthermore, it had large surface area that
carbodiimide hydrochloride (EDC)/N-hydroxysulpho succini- could be tailored to carry multiple PS and additional tumor
mide (NHS) reactions.67b However, PEI is an inherently toxic homing motifs for targeted PDT. It is worth noting that this
polymer. PEGylation, a very often used technique to improve technique is more suitable for loading cationic hydrophilic PSs
water dispersibility of nanoparticle and shield it from being due to the negatively charged nature of the silica matrix.
removed from the circulation by the immune cells, has been However, with hydrophilic PSs, there is a high chance of its
frequently used to modify UCNs.234a,b But one drawback of leakage out of the uncapped pores during systemic circulation.
PEGylation is that it could be a multistep time-consuming This could probably lead to phototoxicity and reduction in the
process. Several PEG block copolymers such as PEG-b-PCL232a amount of PS ultimately reaching the tumor. Despite its merits,
and PEG-b-PLA236 block copolymer have also been employed this method is a slow multistep process and the amount of PS
as polymeric stabilizer. The assembly process is via flash loaded was highly variable from batch to batch. Thus, accurate
nanoprecipitation technique, where all the constituents dosing of the PS becomes a problem, when employing this
including the UCNs and the hydrophobic PS like TPP are technique.
dissolved or suspended in the organic phase, which is then Chemical linkage or covalent conjugation of PS (Figure 18B)
mixed at high speed against a large volume of water, yielding to the surface of UCN offers superior stability, as the PS is
the composite nanoparticle.232a,236 This technique requires chemically attached to UCN surface, and thus could prevent
thorough optimization of the constituents as inadequate PEG leakage of PS during systemic circulation.222a,b,229,234b,238 This
coating will lead to UCN aggregation in physiological solution; method is widely adopted to trap PS to the UCNs as unlike
whereas higher amount of the polymer will lead to formation of chemotherapy drugs, PS need not be released from the UCN
void polymeric micelles. Furthermore, higher polymer concen- construct to exert its cell killing effect. Often PS with a free
tration may lead to attenuation of the UCL. Wang et al. used a carboxyl group (−COOH) is utilized to attach to an amino
PEG-grafted poly(maleic anhydride-alt-1-octadecene) (−NH2) functionalized UCN surface. Qiao et al. utilized a
(C18PMH−PEG) amphiphilic polymer to transfer hydro- slightly different method and grafted HP and SPCD on
phobic UCNs to aqueous phase, yielding PEGylated UCNs mesoporous silica shell through the hydrolysis and condensa-
with excellent water solubility.232b Recently, an outer layer of tion of the silanol groups to achieve stable loading of the PS in
charge-reversible polymer containing dimethylmaleic acid the pores of silica layer.222b In addition to achieving stable
(DMMA) groups and PEG chains were utilized to coat the binding of the PS, this method also allows attachment of
UCNs via a LbL technique involving electrostatic interactions additional functional moieties like targeting ligands to the
in which the outer PEG coated surface is negatively charged at −NH2 group on the UCN surface, enabling targeted delivery to
normal physiological pH 7.4 and switches to a positively cancer cells expressing the receptors to these ligands. PS
charged naked surface at pH 6.8, demonstrating enhanced loading by covalent linkage further offers the advantage of
tumor-homing properties.227b In a different approach, poly- having a short distance between the UCN surface and attached
meric liposomes consisting of amphiphilic polymers of PS molecule, thus maximizing the FRET efficiency between the
octadecyl-quaternized lysine modified chitosan (OQLCS) two. However, despite of the stable binding ability, the PS
functionalized with FA and transactivating-transduction protein payload achieved by covalent linkage was comparatively low,
(TAT) were utilized, which displayed multiple functions as limiting the application of such constructs to in vitro studies.
carriers of UCN and PS, as well as tumor targeting agents while Physical adsorption (Figure 18C) of PS to the UCN surface
imparting good water dispersibility.237 Among biodegradable utilizes noncovalent hydrophobic forces. Usually a hydrophobic
polymers, wrapping of UCNs with positively charged, hydro- PS is adsorbed on to the hydrophobic layer on the UCN. While
philic, and nonantigenic polymer chitosan has shown to be very transferring the bare UCNs capped with a hydrophobic layer
effective in accumulating within the negatively charged tumor (such as an oleic layer) into an aqueous phase by wrapping it
cell membrane due to electrostatic interactions.234 The with polymers like PEI,67b PEG,232,236 chitosan,234a,c α-
amphiphilic nature of chitosan was exploited to harbor cyclodextrin,227a or Tween-20,220 a thin hydrophobic layer is
hydrophobic PSs like Ce6 and PpA, and at the same time created beneath the polymer on the surface of the UCN. The
they can carry various functional groups on the outside for hydrophobic PS attaches to this thin hydrophobic layer
further chemical modifications with other targeting moieties. sandwiched between the UCN and polymer, via hydrophobic
Surface coating of UCNs with a thin layer of silica was found forces. This also allowed close binding of the PS to the UCN
to be stable as silica was resistant to enzymatic or pH core, and hence a better energy transfer from the UCN core to
dependent degradation.230 Silica encapsulation (Figure 18A), the adsorbed PS was achieved. In addition to the superior
which involves incorporation of PS in the silica shell, is a widely energy transfer, this method of PS loading was found to attain
employed strategy to load PS to UCN.226b,230,235 The porosity/ the highest PS payload among the different strategies proposed
pore size of the silica layer can be tuned such that it can host a to date. However, a higher PS loading does not always
wide range of differently sized PS molecules. The presence of guarantee a better PDT efficacy, as at high concentrations the
pores/channels is advantageous to such PDT system, which PS molecules could aggregate or form dimers239 which could
requires the oxygen and ROS to diffuse in and out of the inversely affect its performance as PDT agents. Therefore, it
construct to promote effective cell killing during PDT. The becomes essential to determine the optimal amount of PS that
AC DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Table 3. List of Various UCN Nanoconstructs Synthesized to Date for PDT Applications

AD DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Table 3. continued

AE DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Table 3. continued

can be loaded on to the UCNs to achieve maximum PDT effect 5.3.4.3.1. In Vitro PDT. The very first study reported the use of
while fabricating UCNs for PDT. NaYF4:Yb3+, Er3+ UCNs coated with a thin layer of silica, as
Although not very common, electrostatic interactions delivery agents of hydrophobic PS MC-540.230 The UCN
between the counter charged UCN surface and PS were also construct was also functionalized with a tumor targeting anti-
utilized to load PS close to the UCN surface (Figure 18D). MUC1/episialin antibody and thus demonstrated superior
Wang et al. synthesized oleate ligand free UCN surface by acid targeting toward MCF-7/AZ breast cancer cells. In vitro PDT
treatment, which not only imparted hydrophilicity and was performed by irradiating MCF-7/AZ cells with a NIR light
biocompatibility but also exposed the positively charged Ln3+ source at a wavelength of 974 nm, with an output power of 60
ions on the surface of the UCNs.240 These bare Ln3+ ions were mW for 36 min, following which the cells displayed shrinkage
then utilized for attaching electronegative groups of mono- and membrane damage. However, ROS producing ability of
substituted-β carboxyphthalocyanine zinc (ZnPC−COOH) via this nanoconstruct could have been relatively hindered as the
electrostatic interactions. Such interactions allowed very close PS was physically encapsulated with in the nonporous silica
binding of the PS to the UCN surface owing to the short layer. Since the success of PDT is attributed to the efficient
distance between the donor and acceptor, resulting in a very production and diffusion of the ROS, it is important to modify
efficient energy transfer (96.3%, the highest to date), although the UCN in such a way that the ROS diffused out of the
the drug loading was low (1.1 wt %). nanoparticles effectively. Chatterjee et al. demonstrated the
It is to be noted that none of the above-mentioned strategies ability of PEI-modified NaYF4 nanocrystals loaded with ZnPC,
for the destruction of HT29 human colonic adenocarcinoma
guarantee controllable loading or attachment of PS to the
cells.231 In this work, high PS encapsulation efficiency of 97%
UCN, often compromising the repeatability of PDT results. It
was reported by physical adsorption of nonpolar ZnPC on to
is essential to design robust UCN constructs with controllable
the highly nonpolar UCN core. Furthermore, the effectiveness
and stable PS loading, for optimal and reproducible therapeutic of these FA conjugated nanoconstructs in selective targeting
efficiency. Recently, our group reported a facile method of and destruction of folate receptor (FR) overexpressing HT29
stably and uniformly surface coating PS or photocatalyst TiO2 cells following NIR irradiation at 980 nm for 5 min was
on the surface of silica coated UCN and their application in demonstrated in vitro. However, in addition to being toxic, the
PDT (Figure 18E).241 We demonstrated stable coating of a thin positively charged PEI could stick to any cells in vivo regardless
layer of TiO2 on individual UCNs, to form a well-defined core− of whether it overexpresses FR. It was then assumed that
shell structure with uniform size of about 50 nm. Such a design perhaps the addition of a biocompatible PEG coat could
allowed controllable and uniform PS loading on individual decrease the toxicity and increase the availability of the UCNs
UCNs and completely eliminates the possibility of PS leakage, in circulation. A higher 1O2 production using TPP coencapsu-
thus ensuring significant ROS generation for effective and lated PEGylated UCNs was demonstrated by Ungun et al.232a
consistent PDT results in vitro and in vivo.242 Studies are still Here, the PEG coating improved the biocompatibility and also
underway to optimize the surface properties of UCNs for allowed penetration of oxygen and diffusion of the 1O2
efficient and stable PS loading to achieve maximum 1O2 output. produced. Yet, this study did not validate the PDT efficacy of
5.3.4.3. Application of UCNs in PDT. This section these particles in cells as well as in vivo models. To enhance the
summarizes the recent developments in the application of diffusion of generated ROS, our group synthesized UCN
UCNs for PDT (Table 3). construct modified with a mesoporous silica shell onto which
AF DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 19. (A) Schematic diagram of synthesis of chitosan modified ZnPC loaded UCN. (B) TEM images of ZnPC loaded nanoparticles. (C)
Viability of MCF-7 cells incubated with void and ZnPc-loaded UCNs for different irradiation time intervals (0, 5, 10, and 15 min). Reproduced with
permission from ref 234a. 2012 The Royal Society of Chemistry.

the PS (ZnPC) was loaded by physical entrapment.235c In vitro dose, where the first 5 min of irradiation generated most of the
1
studies on MB49-PSA cells confirmed that a concentration of O2 due to the consumption of dissolved oxygen in the
100 μg mL−1 of ZnPC loaded UCN could cause about 70% cell medium. Apart from its application in PDT, the construct also
death when irradiated at 980 nm using a laser output power of exhibited potential for fluorescence and magnetic resonance
0.5 W for 5 min. In a subsequent paper the molecular effects of imaging (MRI) due to the presence of gadolinium ion (Gd3+)
PDT induced cell death was also discussed and the major mode in the UCN core.
of cell death was confirmed to be apoptosis.235a Despite its Recently, chitosan wrapped UCN loaded with PS showed
advantages, it is to be noted that PS loading in such a system is promising results as PDT agents.234a,b Cui et al. reported
highly variable, leading to batch variations due to difference in loading of ZnPC via hydrophobic interactions achieving one of
pore size that cannot be controlled. Second, due to the the highest PS payloads of 10.8% (Figure 19).234a In vitro PDT
presence of a double layer of silica (first layer of nonporous at 980 nm (output power of 0.6 W for 10 min) using 400 mg
silica to reduce surface defects of UCN core, and a second mL−1 chitosan wrapped ZnPC loaded UCN following 24 h
mesoporous layer), the size of the particles were relatively large incubation killed about 80% of MCF-7 breast cancer cells, while
(∼90 nm). Furthermore, there is an increased chance of cells incubated with void-chitosan wrapped UCN demonstrated
particle growth and aggregation due to the higher diffusion rate more than 80% viability following NIR irradiation. The mode of
at higher temperatures during calcination process. Another cell death was predominantly found to be via apoptosis. In a
group also employed a similar technique to load HP and SPCD different study, Zhou et al. demonstrated PS loading via
onto the mesoporous silica coating, over a CaF2 shell coated chemical linkage of PS−pyropheophorbide A to the amino
UCN.222b However, this time the PS was covalently grafted on group of chitosan that was also tagged with a cyclic RGD
the surface of the UCN. The CaF2 shell reduced surface defects peptide, c(RGDyK).234b Such a covalent conjugation rendered
of the core improving the UCL as well as its biocompatibility. stable PS loading to the UCN as well as retained the
In vitro PDT under 980 nm NIR light demonstrated that a spectroscopic and functional properties of the PS. The presence
strong irradiation dose (2.5 W cm−2) was required to transfer of c(RGDyK) demonstrated targeting abilities to integrin αVβ3
energy from UCN core to the PS molecule, to generate enough expressing U87-MG glioblastoma cancer cells and selective
1
O2 in order to promote efficient cell death. The PDT effect destruction of U87-MG cells upon NIR irradiation. In vitro
was found to be proportional to both concentration and light PDT was performed using 980 nm at 0.5 W cm−2 and different
AG DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 20. (A) Schematic illustration of the mechanism of ROS generation upon irraiation of TiO2−UCN with NIR light for PDT. (B) TEM image
of TiO2−UCN (scale-bar: 50 nm). (C) Fluorescence emission spectrum of 1 mg of TiO2−UCN suspended in 1.5 mL of water, under 980 nm
excitation. (D) ROS production from 0, 2, 4, and 8 mg of TiO2−UCN suspended in 2 mL of water, under 980 nm irradiation (2.16 W cm−2). (E)
Dark-toxicity of TiO2−UCN incubated with cancer cells, OSCC and normal cells, NHF for 6 h.Values are means (n = 3) ± SD *P < 0.05 compared
to control cells without exposure to TiO2−UCN. (c) In vitro PDT using TiO2−UCN on OSCC and NHF cells. Values are means (n = 3) ± SD *P
< 0.05 compared to control cells treated with the respective concentration of TiO2−UCN but without exposure to 980 nm laser (i.e., dark-toxicity
values). Reproduced with permission from ref 241. 2014 The Royal Society of Chemistry.

radiation doses ranging between 75 and 300 J cm−2. It was Following 980 nm NIR irradiation, the photoinduced TiO2
shown that for optimal PDT cell kill, a concentration of 200 ug resulted in the generation of more than one type of ROS
mL−1 and photoirradiation energy greater than 225 J cm−2 was including hydroxyl radicals, superoxide anions, and hydrogen
necessary. peroxide, and their potential in inducing cell death was also
Thereafter, Xu et al. reported synthesis of an anti-cAngptl4 successfully demonstrated in vitro. However, we observed
antibody conjugated nanocomposite consisting of thioglycolic saturation of its cell killing ability, such that the photocatalytic
acid functionalized nitrogen-doped-titanium dioxide (N-doped- killing of nanoparticles did not increase much in a dose-
TiO2) nanoparticles, covalently attached on a NaYF4:Yb3+, dependent manner. This could perhaps have been due to the
Tm3+ UCN core.238a Upon NIR irradiation the UCN core formation of large aggregates in the complex biological
emitted visible light which excited the attached photocatalyst environment, hampering its effective uptake into the cells.
N-doped TiO2 to generate highly reactive electron−hole pair. It Hence in a recent and subsequent study, we further surface
further reacts with water and oxygen to produce ROS leading modified the core−shell TiO2−UCNs with polyethylene glycol
to cancer cell death. However, the synthesized nanocomposites (PEG) to impart stealth properties and stability in terms of its
did not seem to have a uniform size, as controlling the number size, toward making it more conducive for biological
of TiO2 nanoparticles attaching to the UCN surface is not applications.242 PEGylated TiO2−UCNs were found to be
practically possible. The study demonstrated in vitro NIR- both biocompatible and hemocompatible, and demonstrated
triggered drug release and targeted cancer cell killing of dose dependent ROS production and corresponding cell killing
malignant A-5RT3 cells predominantly via apoptosis. Realizing abilities in vitro.
the need for controllable and stable loading of PS, very recently A few other groups also reported synthesis of multifunctional
our group developed a method for uniformly surface coating theranostic UCNs for both imaging and therapy. Zhao et al.
photocatalyst TiO2 on individual UCNs, to achieve well- synthesized small (∼38 nm), size-controllable silica coated
defined core−shell structured nanoparticles (Figure 20).241 Our UCN with PS (tetra substituted carboxy aluminum phthalo-
method has the advantage of stable PS attachment with cyanine, AlC4Pc) covalently linked to the silica shell.222a This
minimal batch variations, preventing any possibility of PS allowed close proximity of the PS to UCN core, demonstrating
leakage, thus ensuring consistent PDT results. Here, the a better PDT efficiency where nearly 40% of the cells treated
photocatalyst TiO2 was stably and uniformly coated using a with 100 ug mL−1 where killed after 5 min of irradiation using a
modified Stöber method, following an electrostatically guided 980 nm laser at a power density of 0.5 W cm−2. Solution based
attachment of a Ti precursor on to the silica coated UCNs studies revealed that the synthesized particle had good water
bearing positively charged amino groups on its surface. dispersibility, 100% entrapment efficiency and no PS leakage.
AH DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 21. (A) Schematic illustration of α-CD surface functionalization procedure and the preparation of PS@UCN complexes and (B) 980 nm NIR
induced PDT using PS@UCNs. SEM image of (C) unmodified UCNs and (D) α-CD-UCNs. (D) Room temperature upconversion emission of
oleic acid capped (OA-UCNs) dispersed in cyclohexane (1 mg mL−1, solid line) and α-CD-UCNs dispersed in water (1 mg mL−1, dashed line),
respectively (inset: their corresponding luminescent photographs under 980 nm NIR excitation, power density: 0.5 W cm−2). (D) Cell viability of A-
549 cells treated with Ce6/DOX@UCN complex (980 nm laser density: 1 W cm−2; irradiation time: 5 min). Error bars were based on triplicated
samples. Reproduced with permission from ref 227a. 2013 Wiley-VCH Verlag GmbH & Co. KGaA.

The presence of Gd3+ ions in the matrix further facilitated both coloaded with a chemotherapeutic agent DOX and Ce6,
T1- and T2-weighted MRI. Chen et al. demonstrated water in induced significantly greater cell-kill compared to treatment
oil strategy to trap positively charged hydrophilic PS MB into using a single modality. Similarly, Wang et al. also utilized Mn
the silica shell.226b No leakage of MB was detected from the doping to synthesize UCNs emitting single-band intense red
UCN construct due to strong electrostatic attraction between light (650−670 nm) UCL.227b The Mn2+-doped UCN were
positively charged MB and the silica shell. The construct also further made water-soluble, by replacing the hydrophobic
had potential for MRI due to the presence of Gd3+ ions in the surface ligands through a ligand exchange reaction using PAA.
core. It is to be noted that majority of the hexagonal-phase They, then utilized LbL self-assembly approach to load the
NaYF4:Yb/Er UCNs developed for PDT typically emitted a negatively charged Ce6 conjugated polymer (PAH-Ce6-
strong green and a weak red fluorescence. However, most of succinic anhydride (SA); Figure 22). The UCN/PAA was
the PSs that were associated with such β−NaYF4:Yb/Er first mixed with oppositely charged PAH to form positively
absorbed at the red region of the spectrum. Apparently, the charged UCN/PAA/PAH, on which the negatively charged
dominant green emission has no contribution in the excitation PAH-Ce6-SA was attached via electrostatic interaction, to load
of PS, which resulted in relative low efficiency in the FRET up to 3 layers of the Ce6 conjugated polymer. This technique
process. Recognizing this fact, Tian and co-workers synthesized resulted in achieving a very high loading of PS (up to 11.0 wt %
strong dark red-light emitting α-cyclodextrin (α-CD) modified for 3 layered Ce6 polymer-UCN composites), however 7.7 wt
manganese ions (Mn2+) doped UCNs (Figure 21).227a The % (two-layer) was found to be optimal. The tight coupling
cube shaped UCNs with a single-band red UCL emission between PS molecules and UCNs prevented Ce6 leakage from
peaking at 650−670 nm were further loaded with lipophilic UCNs up to 2 d under physiological conditions. To further
PSs, Ce6 or ZnPc, and hydrophilic MB. As the loading of the impart UCNs with pH-responsive charge reversibility, PAH was
PS on the UCNs employed hydrophobic interactions, the more cografted with PEG and DMMA to obtain negatively charged
hydrophobic PSs (Ce6 and ZnPC) were more efficiently loaded PAH-DMMA-PEG, which was then used to coat UCNs by
compared to MB. In vitro PDT with Mn2+-doped UCNs electrostatic interactions. On incubating the pH-sensitive
AI DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 22. (A) Schematic showing the fabrication process of pH-sensitive charge-reversible UCNs with multilayers of Ce6 loading. (B) TEM
micrograph of Mn2+-doped UCNs; inset: high-resolution TEM image of a UCN. (C) UCL emission spectrum of Mn2+-doped UCNs under 980 nm
laser excitation; inset: photo of a UCN solution under ambient light and under 980 nm laser excitation. (D) T1-weighted MR images of UCN
solutions at different concentrations. (E) T1 relaxation rates (r1) of UCN solutions at different Mn2+ concentrations. (F) Cell viability of HeLa cells
after various treatments indicated with and without the 980 nm laser irradiation as evaluated by the standard MTT assay. Error bars are based on four
parallel samples. Reproduced with permission from ref 227b. 2013 Wiley-VCH Verlag GmbH & Co. KGaA.

UCNs with HeLa cells grown at pH 6.8 and 7.4, significantly dose-dependent decline in the viability of HeLa cells upon NIR
greater UCL signals were obtained from HeLa cell at pH 6.8, irradiation at an intensity of 1.37 W cm−2 confirmed the
indicating significantly enhanced cellular internalization at capabilities of this multifunctional UCN−fullerene nanoplat-
slightly acidic pH due to their charge conversion from negative form in NIR imaging guided cancer therapy. Recently, Chen
to positive. This also led to a correspondingly high cell death and co-workers demonstrated for the first time synergistic
following irradiation at 980 nm light. In addition to the PDT/PTT photokilling effects of bovine serum albumin (BSA)
enhanced therapeutic effect, the Mn2+ doped nanoconstruct modified NaGdY4-based UCNs.256 Here, two types of dye
also demonstrated potential as T1-weighted contrast agent in molecules, including a PS, RB, and an NIR-absorbing dye,
MR imaging. In order to develop a T2-weighted MRI contrast IR825, were simultaneously loaded into the BSA layer via
agent, Zeng et al. reported the synthesis of multifunctional hydrophobic interaction, so that under 980 nm excitation RB
MRI/UCL/PDT nanocomplexes composed of 10 nm Fe3O4 absorbs green light emitted from UCNs and induced
core, coated with a tetragonal NaYF4:Yb/Er shell.252 The photodynamic cancer cell kill, while IR825 under 808 nm
nanoconstruct also exhibited good therapeutic performance laser irradiation offers a strong photothermal effect. Although
which resulted in nearly 70% MCF-7 cell death following the PTT effect (808 nm excitation, 0.5 W cm−2 for 5 min)
irradiation at 980 nm at a power density of 0.02 W cm−2 for 5 caused by the hyperthermia induced by IR825 loaded
min. A different approach was adopted by Liu and co-workers, nanocomposites was slightly more effective in killing 4TI cells
where they fabricated UCNs by separately doping core and in vitro than the PDT effect (980 nm excitation, 0.4 W cm−2 for
shell with Er3+ and Tm3+ instead of homogeneous codoping, to 10 min), the combination of both therapies induced a
achieve strong simultaneous multicolour UCL around 450, 475, synergistic effect resulting in significantly greater cell kill
540, 650, and 808 nm upon 980 nm NIR excitation.229 They compared to the monotherapy.
then utilized monomalonic fullerenes (C60MA), covalently 5.3.4.3.2. In Vivo PDT. The biggest challenge in the application
conjugated to the core−shell structured UCNs as energy of UCNs for PDT in vivo has been the delivery of the particles
acceptors and ROS generating agents. The nanocomposites such that it selectively accumulates in the tumor, preventing
were additionally modified with PEG-succinimidyl carbonate systemic toxicity. Most of the earlier studies reported delivery
(PEG-SC), to improve biocompatibility and stability in of UCNs via intratumoral injections. This route of admin-
biological solvents and further conjugated with FA to bestow istration was chosen as the particles synthesized initially were
targeting abilities to folate receptor expressing HeLa cells. A large and aggregated in the physiological milieu. Furthermore, it
AJ DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Table 4. Summary of Various PDT Parameters for in Vivo Application of UCNs

AK DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 23. (A) Schematic drawing of NIR-induced PDT using UCN-Ce6. (B) TEM image of PEGylated UCNs. (C) Growth of 4T1 tumors on
different groups of mice after various treatments indicated; relative tumor volumes were normalized to their initial size (n = 10). (D) The survival
curves of mice in 60 days after various treatments indicated; UCN-Ce6 PDT treatment drastically prolonged survival time of mice. (E) Schematic of
in vivo PDT experiment where tumors were hidden below a slice of pork tissue (8 mm) during light treatment. (F) The growth of 4T1 tumors on
different groups of mice after various treatments indicated. The relative tumor volumes were normalized to their initial sizes. (n = 10) *p < 0.05.
Error bars represent standard errors of the mean. Reproduced with permission from ref 232b. 2011 Elsevier.

had limited specificity and selectivity to the tumor that most of the animals. Furthermore, there was an enhanced retention of
them accumulated in the organs of the MPS leading to toxicity. the UCNs in the organs such as spleen and the liver 15 days
Thus, to obtain higher PDT efficiency and fewer side effects, post injection, indicating apparent translocation of the UCNs
UCNs were administered intratumorally. The various in vivo from the injected site via the lymphatic circulation. In vivo PDT
studies and the parameters used in PDT are summarized in using 980 nm laser irradiated at a power density of 0.5 W cm−2
Table 4. for 30 min (1 min interval after each min of irradiation)
Wang et al. demonstrated in vivo PDT in murine breast appeared quite promising with 70% of the tumors disappearing
tumor using Ce6 loaded PEGylated UCNs (Ce6-PEG-UCN; without regrowth in 2 weeks, exhibiting longer morbidity free
Figure 23).232b PEGylation improved the stability as well as survival. It was clearly shown that NIR light penetrated the 8
decreased the toxicity of Ce6-PEG-UCN, where the hydro- mm pork tissue placed over the tumor, triggering Ce6-UCN in
phobic Ce6 molecule was simply adsorbed onto the oleic layer the tumor leading to significant reduction in tumor growth rate.
of the PEGylated UCN, to achieve a very close binding of Ce6 However, direct application of NIR light to the tumor did not
on the UCN. This strategy had an added advantage of offering generate as much 1O2 as that generated by free-Ce6 triggered
a higher resonance energy transfer between the photon donor by visible 665 nm light, thus suggesting that NIR-induced PDT
and acceptor molecule, by virtue of its adjacency. The size of using UCN has the potential to be developed for the ablation of
this particle (∼30 nm) was the smallest compared to all the solid, deep seated tumors.
previously reported UCNs for PDT. Yet, particles were Cui et al. reported in vivo PDT using chitosan modified
administered intratumorally, rather than intravenously into UCN loaded with ZnPC.234a PDT at 980 nm NIR laser
AL DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 24. (A) Schematic diagram of ZnPC and MC540 coloaded with in the mesoporous silica shell coated UCN. (B) TEM image of the
mesoporous-silica-coated UCNs (scale bar, 50 nm). (C) Schematic diagram showing UCN-based targeted PDT in a mouse melanoma model
intravenously injected with UCNs surface modified with FA and PEG. Scale bar, 10 mm. (D) Change in tumor size as a function of time following
intravenous injection of UCNs with and without targeting agent (FA) to assess the effectiveness of UCN-based targeted PDT in tumor-bearing mice.
Values are means ± s.e.m. (n = 7 mice per group). *P < 0.05 compared to control group 3 by Kruskal−Wallis ANOVA. (E) Representative images of
mouse intravenously injected with FA-PEG-UCNs, unmodified UCNs or PBS showing the change in tumor size (highlighted by dashed white
circles) before (0 d) and 7 d after PDT treatment. Scale bars, 10 mm. Reproduced with permission from ref 235b. 2012 Nature Publishing Group.

irradiation at 0.4 W cm−2 for 15 min, following intratumoral the emitted upconverted light was being utilized for maximum
administration of these particles produced significant reduction PDT efficacy. The results were quite promising in that all the
in tumor via necrotic cell death, within 14 days compared to the three modes of UCN administration produced significant
untreated group. In addition, there was no sign of adverse side- reduction in tumor volume compared to the untreated group.
effects and lethality among PDT treated animals due to the However, a very long irradiation time of 1−2 h (at 980 nm,
presence of a nontoxic and biocompatible chitosan coat. power density of 415 mW cm−2), was employed for in vivo
The first study to demonstrate systemic administration of a PDT. This could have been due to the inefficient energy
42 nm UCN carrying Ce6 was by Park et al.223 Here, Ce6 was transfer from UCNs to the PS probably due to the limited PS
attached both via physical adsorption to phospholipid layer as loading capacity (<0.6 wt %) and poor stability of the PS that
well as covalent conjugation to the amino group on UCN, was physically adsorbed onto the mesoporous silica. Perhaps,
achieving stable and highest PS payload (>1000 Ce6 covalent binding of the PS to the surface of UCNs could
molecules/UCN). Due to the presence of the biocompatible enhance the upconversion efficiency and produce a more
polymer PEG, these UCNs carrying Ce6 had better circulation pronounced PDT effect in vivo.
half-life than free-Ce6. Particles mainly accumulated in the liver Cui et al. again demonstrated remarkable therapeutic efficacy
and spleen; however the amount dropped considerably 7-days with tumor inhibition ratio of up to 50% using FA conjugated
post injection, reducing long-term toxic effects. Following in chitosan modified ZnPC loaded UCNs in comparison with a
vivo PDT at 980 nm, there was a 6-fold decrease in tumor size tumor inhibition ratio of 18% achieved by conventional visible
among the treated group, thus clearly demonstrating its light-activated PDT.234c Here, PS was loaded via hydrophobic
potential as PDT agent. Thereafter, our team reported interactions to achieve high drug loading capacity of about 10%.
therapeutic efficiency of mesoporous silica coated UCNs The FA conjugated UCNs were administered intravenously and
coloaded with two different PS, ZnPC and MC-540 (Figure exhibited high tumor targeting efficiencies in vivo, where it
24), using mouse melanoma model in a stepwise fashion.235b accumulated as early as 4 h post administration, whereas the
First, the melanoma cells were prelabeled with PS loaded unconjugated UCNs became visible only after 12 h. The
UCNs and then injected subcutaneously to develop UCN subcutaneous tumor developed on the mouse were covered
loaded tumors in mice. This was followed by assessment of with 1 cm pork tissue to simulate tumors inside the body and
PDT efficacy by injecting PS loaded UCNs intratumorally into was irradiated with either 660 or 980 nm light following
tumor bearing mice and subsequently intravenous admin- administration of PS loaded UCNs. Survival rates of mice in
istration of FA conjugated- PEGylated- PS loaded UCNs was this deep penetrating PDT experiments reached 88% for 980
performed. This was the first time that two different PSs nm induced PDT compared to 75% using 660 nm light
absorbing at two different wavelengths (660 and 540 nm) of irradiation demonstrating the ability of the nanoconstructs in
AM DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

the ablation of deep-seated tumors. The results indicated that


PDT treatment based on FASOC-UCN-ZnPc could effectively
improve the survival quality of mice and prolong their lifetime.
Using the pH-sensitive charge reversible nanoconstruct, Wang
et al. demonstrated significantly delay in tumor growth
following 980 nm NIR PDT at 0.5 W cm−2 for 30 min (900
J cm−2), 1 day after intratumoral injection of 0.4 mg UCNs per
4T1 tumor-bearing Balb/c mouse.227b At the same time,
significantly higher uptake of UCNs (6.4 folds) were observed
in the tumor, 2 h following intravenous administration of same
dose of UCNs. Despite an increased tumor accumulation of
UCNs, the majority of injected dose were observed in
reticuloendothelial systems (RES), such as the liver and spleen,
following systemic administration. This warrants further
improvement of the surface modification techniques, to
enhance the tumor targeting, and at the same time, reduce
the accumulation and retention of these smart UCNs in the
RES. In their recent publication, Chen et al. demonstrated
synergistic therapeutic efficacies of the combined PTT and
PDT in an in vivo mouse model bearing 4T1 tumors, using a
double dye loaded (RB and IR825) BSA modified UCNs.256
The combination therapy (PDT at 980 nm excitation for 30
min at 0.4 W cm−2 followed by PTT at 808 nm, at 0.5 W cm−2
for 5 min) after intratumoral injection of 10 mg mL−1
RB:IR825 coloaded UCNs significantly inhibited tumor growth
in vivo, while those after monotherapy (PDT only or PTT
only) showed a delayed tumor growth. In addition to the
therapeutic effect, intrinsic paramagnetic and optical properties
of Gd3+-doped UCNs were utilized for in vivo dual modal
imaging, thus proving to be excellent multifunctional Figure 25. (A) Schematic illustration of multifunctional LiYF4:Yb/Er
theranostic agents. UCNs for cancer theranostics. Variation of (B) tumor volumes and
(C) body weights of mice in experiment and control groups,
After successfully developing a positively charged ligand free respectively. Each data point represents the average value of 5 mice.
UCNs on which electronegative PS were tightly coupled via (D) Representative photos of a mouse showing tumors at 14 days after
electrostatic interactions, to obtain a UCN-PS complex with treatment in experiment and control groups, respectively. (E) Images
highest FRET efficiency (96.3%) until date, Wang et al. studied (left) and the corresponding high-resolution images (right) of H&E
the in vivo therapeutic efficiency of the developed PVP coated stained tumor tissues harvested from the experiment and control
UCNs, in a H22 hepatocarcinoma mouse model (Figure groups after 14 days. Reproduced with permission from ref 240. 2014
25).240 NIR PDT of the tumor at 980 nm, 12 h post Royal Society of Chemistry.
intratumoral injection of 20 mg mL−1 ZnPC−COOH loaded
UCNs exhibited significant delay in tumor growth (501 mm3) linkage, minimized premature leakage and improved its
compared to untreated control (1282 mm3) by 14 days. bioavailability. In vivo PDT in mice models of tumors following
Compared to the animals in the untreated control, the body intratumoral delivery of these ALA loaded nanoconstructs
weight of the mice in the treatment group remained unchanged, demonstrated significant tumor growth delay compared to
indicating an overall improvement in their health status. In our controls even in the presence of 12 mm thick pork tissue; the
recent study in which we directly surface coated photocatalyst greatest depth at which significant reduction in tumor growth
TiO2 on individual UCN core and further surface modified was achieved with UCN-PDT.
them with PEG to improve its biocompatibility, we The first ever attempt that combined bimodal imaging and
demonstrated remarkable delay in tumor growth and significant trimodal therapy in a single UCN-based nanotheranostic
improvement in survival of the animals in the treatment groups system was yet recently reported by Fan et al.264 They
compared to the control groups (Figure 26).242 It can be synthesized a sub-80 nm rattle structured nanocomposite with
concluded that stable and controllable loading of PS is essential Gd3+ doped UCN in the core (for simultaneous UCL and MR
for consistent and optimal PDT results. imaging), and a mesoporous silica as the outer shell with a
In order to improve the UCL emission in the red region for cavity in between them (UCMSNs; Figure 28). HP that serves
further improvement in the penetration depth achievable by as both PS and radiosensitizer was covalently grafted inside the
UCN-based PDT, recently Punjabi et al. developed CaF2 silica shell with a loading efficiency of 7.8%, whereas the
coated α-NaYF4:Yb,Er in which the Yb doping was increased chemo-drug docetaxel (Dtxl) was encapsulated with in the
from 20% to 98% (where 80% Yb doping showed optimal inner cavity with a loading efficiency of 4.5 wt %. The
results; Figure 27).266 They reported 15-fold increase in red- nanoconstruct was found to accumulate in the tumor by passive
emissions compared to their hexagonal phased counterparts (β- targeting with a 25% enhancement in T1-weighted MRI signals
NaYF4:Yb(20%),Er(2%)@β-NaYF4), with an absolute quan- within 15 min post intravenous delivery. By simultaneously
tum yield of α-NaYF4:Yb(80%),Er(2%)@CaF2 measured to be imposing NIR excitation and high energy X-ray irradiation
3.2%, the highest reported value for red-emissions. Further- following intratumoral administration of UCMSNs-HP-Dtxl,
more, conjugation of ALA to the UCN surface via a hydrazone the tumors in mouse models were quickly eradicated due to the
AN DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 26. (A) In vivo OSCC tumor growth up to 35 days on different groups of mice after various treatments indicated, n = 5 per group. Error bars
represent standard error of the mean. Representative images of animals with tumors that were (B) not treated (untreated control animals) and (C)
intratumorally injected with 0.1 mg Mal-PEG-TiO2−UCN and irradiated with 980 nm light, at designated time-points. (D) Survival rates of mice in
different treatment groups within 60 days. Reproduced with permission from ref 242. 2014 American Chemical Society.

synergistic effect of PDT/chemo-/radiotherapy. However, feeding C. elegans with UCNs did not affect its life span, egg
following intravenous delivery and trimodal therapy, the tumors production, egg viability, or growth rate, indicating that UCNs
were found to regrow after an initial drop in the volume. It is to were not significantly toxic except at concentrations above 5 mg
be noted that most of the in vivo tumor regression studies were mL−1 or higher.268 A list of all the published efforts on
short-term (10−14 days), where majority of the results report a biodistribution, toxicity, and clearance of various UCN
delayed tumor growth and improved overall survival, rather constructs in animals are summarized in Table 5.
than a complete tumor regression. To further improve the To gain insight into the kinetics of these nanocrystals in
therapeutic efficiency of UCN based nanoconstruct and fine- rodents, Jalil et al. studied the biodistribution of silica coated
tune the PDT regimen to achieve complete or partial tumor UCNs following intravenous injection in Wistar rats, up to 7
regression, long-term tumor regression studies need to be done. days.269 No obvious acute toxic or adverse health effects were
5.3.4.4. Toxicity and Biodistribution of UCNs. Although observed at the dose level of 10 mg kg−1 for up to 1 week post
UCNs have been proposed for various bioimaging and administration. Quantification of yttrium content in various
therapeutic applications, numerous unresolved issues with organs at specfic time-points revealed that lung and the heart
respect to its interaction with biological system yet remains. were the main target organs where the nanocrystals
Like many other inorganic nanoparticles, UCNs are not easily accumulated immediately (29.2 and 18.0 mg L−1 per g of
biodegraded in the biological system, causing concerns about its tissue at 10 min) post injection. However, the amount dropped
long-term toxicity and side effects. Although, humans are significantly by 24 h. The nanoparticles were also detected in
exposed to lanthanides based compounds in the form of the spleen, reaching the highest concentration at 30 min (6.3
therapeutics and diagnostics, they are often considered to be mg L−1 per g) as well as in the kidneys, where the
moderately to highly toxic. Generally, the toxicity depends on concentration of yttrium remained the same up to 24 h.
the compound (inorganic versus chelated compounds), route However, the concentration in blood and liver remained low
of administration, and dose.267 Thus, the behavior, biodis- throughout. The nanocrystals were mostly cleared from all
tribution, degradation, clearance, and toxicology profiles of major organs by day 7 postinjection indicating that they were
UCN in biological systems are the most critical and not retained for a long period of time. On studying the long-
fundamental question to be addressed presently. Almost all term fate of 11.5 nm sized poly(acrylic acid) (PA) coated
studies highlighted in this review paper, have evaluated the in UCNs in nude mice, Xiong et al. reported that the major organs
vitro cytotoxicity of respective UCN constructs by the standard that retained the nanoparticles were the liver and spleen
CCK-8, MTT, MTS, and LDH assays, and have found to be (Figure 29).270 Although, no major toxic effects were observed,
safe to a broad range of tested cell lines within a certain the study reported a very slow clearance of the nanoconstructs
concentrations (usually in μg mL−1 range) and a limited from the system via hepatobiliary route. However, the study
incubation period (usually ranging from 30 min to 24 h). A few only included a small number of animals (n = 3) and the blood
studies utilized Caenorhabditis elegans (C. elegan) as a model biochemical and histology results were collected only at the end
organism for evaluating the toxicity of UCNs and reported that of the experimental period (day 115).
AO DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 27. (A) Emission spectra under 980 nm, 1 W cm−2 excitation of α-NaYF4:Yb,Er@CaF2 UCNs with different Yb-levels. (B) Integrated counts
of red-emission and photographs (inset) of α-NaYF4:Yb,Er@CaF2 UCNs with different Yb-levels. (C) TEM images of PAA−UCNs (a), hydrazide-
functionalized UCNs (b), and ALA−UCNs (c). (D) In vivo volume of tumors exposed to various controls and ALA−UCNs with red and near-
infrared irradiation (0.5 W cm−2) in simulated deep tumors. Statistical significance was determined from one-way t tests; significance (*) was based
on p < 0.05 and p > 0.05 for not significant (**) pairs. Reproduced with permission from ref 266. 2014 American Chemical Society.

Later, Cheng et al. conducted a thorough assessment of the studies could have led to the observed difference in the
pharmacokinetics, long-term biodistribution and toxicity of 30 excretion routes and clearance of the nanoparticles. But it is to
nm sized PEG or PA modified UCNs.271 Although, the blood be noted that, although the nanoparticles remained in the
circulation curves of UCN-PEG and UCN-PA followed a two- animal for over 90 days, no appreciable variations were
compartmental model, the blood circulation half-life of UCN- observed in the blood biochemical, hematological or histo-
PEG was significantly greater than UCN-PA, which was pathological analysis.
attributed to the biocompatible PEG coating on the UCN Presence of targeting ligands were found to channel the
surface and its improved stability in physiological solutions. In UCNs to the tumor and reduce the percentage of injected dose
vivo UCL imaging showed that UCN-PA accumulated in liver that accumulated in other organs such as the liver and spleen.
within 5 min, but UCN-PEG uptake was slower and observed Cui et al. administered FA conjugated UCNs intravenously,
in liver at 30 min. Ex vivo UCL imaging revealed accumulation that exhibited high tumor targeting efficiencies and accumu-
of UCNs in the liver, spleen, lung and bone at 1 day, but no lated in the tumor as early as 4 h post administration, whereas
signal was detected by day 7. However, the yttrium ion levels in the unconjugated UCNs became visible only after 12 h.234c The
these organs did not drop much over a course of 90 days, particles first accumulated in the liver, following which they
except in the lung, which showed a rapid decrease of yttrium were gradually excreted into the intestines by 24 h and the
ion concentration after 3 days. The presence of nanoparticle absence of fluorescence signals from the loaded NIR dye (ICG-
aggregates in the TEM of liver slices from mice 7 days post Der-01) at 48 h indicated complete clearance of the UCNs
injection, indicates partial decomposition of UCNs inside the from the system via enterohepatic route. A significant variation
macrophage cells after RES uptake and loss of their UCL in the biodistribution was observed in the presence of a
emission signals over time. Thus, the study concluded that targeting moiety FA at 24 h in a tumor bearing mice, where FA-
nanoparticle residues would stay inside the mouse body for conjugated UCN levels were much higher in tumor but lower
more than 90 days, which is in stark contrast to the previous in organs like lung, liver, and kidney, compared to the level of
reports. Perhaps the size difference of UCNs in these two unmodified UCNs. Due to the huge diversity in the nature, size,
AP DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 28. (A) Schematic illustration of the synthetic procedure of UCMSNs. Gd-UCNPs were prepared by epitaxial growth NaGdF4 layer on
NaYF4:Yb/Er/Tm through a typical thermal decomposition process. A dense silica layer was then coated on Gd-UCNPs by a reverse microemulsion
method, designed as Gd-UCNPs@SiO2. Subsequently, a mesoporous silica shell was deposited on Gd-UCNPs@SiO2 via the template of CTAC,
designed as Gd-UCNPs@SiO2@mSiO2. Finally, UCMSNs were successfully fabricated based on a “surface-protected hot water etching” strategy. (B)
Transmission electron microscopic (TEM) images of (a) Gd-UCNPs (NaYF4:Yb/Er/Tm@NaGdF4), (b) Gd-UCNPs@SiO2, (c) Gd-UCNPs@
SiO2@mSiO2, and (d) UCMSNs (scale bar = 50 nm) (C) In vivo T1−MRI images of a 4T1-tumor bearing mouse after intravenous injection of
UCMSNs at designated time points: (a) 0 min; (b) 5 min; (c) 10 min; (d) 15 min. (e) Comparison of MRI signal intensity of the corresponding
4T1-tumor at different time points from 0 to 15 min. (f−h) Ex vivo NIR−NIR upconversion luminescent imaging (UCL) of dissected organs after
the intravenous injection of UCMSNs: 1, heart; 2, liver; 3, spleen; 4, lung; 5, kidney; 6, tumor (i−k). (D) Tumor growth and relative tumor volumes
of different groups post intratumoral injection of UCMSNs-HP-Dtxl and corresponding treatment. Control groups received PBS. (E) Tumor growth
and relative tumor volumes of different groups post intravenous injection of UCMSNs-HP-Dtxl and corresponding treatment. Reproduced with
permission from ref 264. 2014 Elsevier.

composition, charge, surface properties, and presence or UCNs, it becomes very hard to even speculate the
absence of additional functional moieties on the surface of biodistribution and toxicity of these nanomaterials, with the
AQ DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Table 5. Summary of the Published Efforts on Biodistribution and Clearance of UCNs in Animal Models

limited knowledge available from the literature to date. Several particular organ still remain unanswered. Long-term compara-
questions regarding the ultimate fate of these nanomaterials in tive studies on the biodistribution, metabolism, degradation,
the system, the possibility of their degradation into toxic and toxicity profiles of various UCNs of different size, charge,
byproducts, and their effects following prolonged residence in a and surface functionalities are necessary to evaluate the
AR DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 29. (A) Real-time in vivo UCL imaging of athymic nude mice after intravenous injection of PA-UCNs (15 mg kg−1) at different time points.
Column 3: overlays of UCL and brightfield images of mice. Column 6: overlays of UCL and brightfield images of dissected mice. (B) Biodistribution
of particles in organs of mice. Error bars were based on triplet measurements. (C) H&E-stained tissue sections from mice 115 days postinjection
with PA-UCNs (a, c, e, g, i, and k) and mice receiving no injection (b, d, f, h, j, and l). Tissues were harvested from heart (a, b), spleen (c, d), liver (e,
f), lung (g, h), kidney (i, j) and blood smear (k, l). Reproduced with permission from ref 270. 2010 Elsevier.

usefulness of this class of nanomaterials before its further lagging behind is a thorough understanding of how they behave
development and application in the clinics. as a collective entity and affect complex biological systems. In
this regard, there is an urgent need for in-depth characterization
6. CONCLUSION AND FUTURE PERSPECTIVES of such multifunctional therapeutic agents to check if they
cross-talk, quench, or interfere with each other’s capabilities
PDT is undoubtedly a promising therapeutic option in the impeding the overall outcome. In addition to that, standardized
management of cancer. However, it has still not gained physicochemical characterization, validation, and safety proto-
acceptance as a first line treatment option mainly due to the cols must be established to achieve pure and homogeneous
shortcomings of classical PSs. The application of nanoparticles nanoparticles that can yield precise and reproducible results.
in the field of PDT is an extremely promising avenue for future Furthermore, the fact that none of such multifunctional
technological breakthroughs. Nanoparticles can act as carriers theranostic agents have been approved by FDA for clinical
of hydrophobic PSs and transport high PS “payload” to the application is undeniably worrying. Again, the major bottleneck
tumor site via the EPR effect. Additional surface modification for the translation of these theranostic agents into clinical
and functionalization with targeting moieties could further practice is the absence of standardized procedures to assess its
enhance the selective accumulation of the PS loaded nano- in vivo biodistribution, detailed pharmacokinetic pharmacody-
particles at the target site. In recent years, much effort has been namic (PK−PD) analysis, short-term and long-term effects of
channelled in the development of multifunctional theranostic the nanomaterials in vivo, as well as its clearance from the
agents where a number of diverse diagnostic and therapeutic system. This was exactly the reason behind the establishment of
functionalities have been incorporated in a single nanoplatform National Characterization Laboratory by the U.S. National
for a “see and treat” approach. The versatility of such agents Cancer Institute (NCI) in 2004, which has developed
allows delivery of drugs to the diseased/target site with standardized evaluation protocols for rigorous characterization
enhanced spatial specificity while achieving simultaneous real- of nanoparticle’s physicochemical properties, in vitro immuno-
time imaging, monitoring, as well as therapeutic capabilities. logical and cytotoxic characteristics, as well as administration,
While, an in-depth knowledge and understanding of nanoma- distribution, metabolism, and elimination (ADME)/toxicity
terial chemistry, nanoscale manufacturing and synthesis has profiles in animal models; to facilitate the regulatory review of
evolved tremendously in order to develop novel nanoplatform nanotechnology-based cancer therapeutics and fast track the
incorporated with sophisticated functionalities, what is now clinical trial process. Today, a decade later, more than 300
AS DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

nanoparticle based cancer therapeutics have been characterized being carried out at the solution- and cell-based level. Solution-
with many more in the pipeline. More such regulatory agencies based studies help to predict nanoparticle toxicity by assessing
that provide a “one-stop” solution for systematic step-by-step the particle size, size distribution, morphology, composition,
characterization of nanomaterial need to be established, given dispersion, surface area, surface chemistry, and surface
the vast number of promising efforts from both academia and reactivity. It is to be noted that nanoparticles behave differently
the biopharmaceutical industry. in different solutions and its dispersion in solutions rarely
In this review, we have provided a comprehensive account on reflect its distribution at the primary particle size. Nanoparticle
the fascinating development of various nanoparticles and their agglomeration could lead to size-, surface area-, and dose-
application in PDT. As most of the classical PSs have dependent toxicity.277 Furthermore, it raises concerns about
limitations such as poor solubility under physiological efficiency of ROS production due to the altered photophysical
conditions, undesirable pharmacokinetics, low tumor selectivity, characteristics of the agglomerated PS loaded UCNs. Hence, it
a suitable nanoplatform carrying the PSs is expected to resolve is highly essential to study its dispersion characteristics and
these problems to a certain extent. Among the various stability in physiologically relevant buffers over a period of time,
nanoconstructs, polymeric (synthetic or natural), liposomal, before its characterization in vitro. However, this is hardly
or dendrimeric nanoparticles are biocompatible and biodegrad- performed in any study until date, perhaps due to which its
able, and hence have the added advantage of being enzymati- efficacy seem to be undermined under in vitro and in vivo
cally hydrolyzed and excreted from the body, minimizing long- conditions. A better knowledge of the characteristics of UCNs
term accumulation.66 These nanoparticles are very versatile due at conditions close to the physiological setting will further
to the existence of a variety of polymers and manufacturing streamline its optimization very early during the evolution of
methods, and thus the chemical composition and architecture the construct and steer its development as a better PDT agent.
of the nanoparticles can be customized to accommodate PSs For effective PDT, optimization of the PDT parameters such
with varying degrees of hydrophobicity, molecular weight, or as the DLI, laser dosimetry including the fluence, fluence rate,
charge.67b,274 Furthermore, the surface properties, morpholo- and time of irradiation is necessary. For PDT using UCNs, a
gies, and compositions of polymeric matrices can be easily laser power density ranging between 0.4 W cm−2 to 134 W
optimized to achieve controlled degradation of the polymer and cm−2 and fluence >1000J cm−2 have been typically used.
drug release kinetics.275 While these nanoparticles can be However, in clinical application and PDT experiments using
tailored to carry multiple functionalities such as targeting conventional PS, power density <1 W cm−2 and fluence <1000
moieties or additional imaging and therapeutic capabilities, J cm−2 will probably be enough for efficient tumor cell death.278
nonbiodegradable nanoparticles provide in-built multifunc- Apparently, UCN based PDT works via anti-Stokes emission
tional capabilities due to its intrinsic properties. Some process that is reported to have a quantum yield of less than 1%
nanoparticles such as fullerenes, TiO2, and ZnO can also be in general.279 Hence, the efficiency of this process is less than
excited by light of a specific wavelength to induce generation of direct excitation of PS by visible light source in conventional
ROS. A third type can transduce energy to the accompanying PDT. Coating of an undoped shell (such as NaYF4 or CaF2)
PS via FRET, in order to excite PS indirectly at wavelengths around the UCN core have shown to improve quantum yield
that it does not normally absorb, such as X-ray nanoparticles, by the suppression of surface quenching effects.265 The low
QDs and gold nanorods. Although TP-PDT allows deep-tissue quantum yield also results in unsatisfactory brightness of the
penetration via simultaneous excitation with two low-energy UCNs under low excitation power densities. The brightness of
NIR photons, the treatment is restricted to a tiny tissue volume the UCN is dependent on the 4f-4f optical transition
resulting in prolonged treatment time, thus limiting its clinical probabilities of the lanthanide ions, which are remarkably
use. On the other hand, PDT using UCNs doped with PS is affected by their local crystal field.280 Consequently, fine-tuning
becoming the new sensation in the field with clear advantages the local crystal field of the lanthanide ions is an effective
over most other nanoparticles. UCNs act both as carriers of PS strategy to increase the UCL of lanthanide ion-doped
as well as enables indirect excitation of the accompanying PS nanocrystals.281 Doping the UCNs with other elements or
with upconverted light upon excitation with low energy NIR dyes with strong absorption and further optimization of the
light. Recently, there has been a growth in the amount of doping ion ratios,282 varying the crystal phase of the
literature available in this area which clearly depicts the growing nanocrystals,283 fabricating core−shell structures,284 and
interest, clear advantages, and possibility of further develop- utilizing metal-enhanced fluorescence285 are among the
ments in the field. However, long multistep reaction methods methods being explored to improve the brightness and
associated with UCN synthesis and PS loading often leads to quantum yield. Currently, much of the research in this field
batch to batch differences, making it difficult to compare uses an NIR excitation wavelength of 980 nm that matches the
results. This is because even subtle changes in composition absorption of the commonly utilized sensitizer Yb3+. However,
arising from minute deviations in the manufacturing process water has strong absorption around 970 nm, which could lead
could substantially affect the PK, biodistribution, and toxicity in to heating up of biological tissues when excited around this
vivo.276 Thus, stringent quality control measures in terms of wavelength. In addition to the heating effects, a significant
uniformity in particle composition, size, surface chemistry, PS reduction in the penetration depth is anticipated as the 980 nm
loading, etc. are necessary for commercialization of this light gets attenuated, while traveling through tissues. A shift in
technology. Further simplification of the synthesis steps, excitation wavelength from 980 nm to a lower NIR wavelength
standardization of nanoparticle preparation, testing and that does not cause a thermal effect could be more clinically
validation protocols, utilization of automated nanoparticle appropriate. Efforts to excite UCNs using another absorption
fabrication processes while implementing good manufacturing peak of Yb3+ at 915 nm were accompanied by a reduction in
practices would allow greater consistency in the quality of the excitation efficiency.286 As an alternative, NIR dye sensitized
UCNs synthesized, besides enabling easy scaling up for UCNs were reported to harvest light in the shorter NIR
commercialization. Presently, much of the research is still wavelengths (740 to 850 nm), where the surface bound NIR
AT DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

dye served as an antenna that absorbed light in the specified field, it is imperative to have frequent facilitations between
spectrum enabling FRET to UCN core to produce an scientists, academicians, engineers, clinicians, and industrial
upconverted light.287 However, such NIR absorbing organic innovators to share and discuss the opportunities and
dyes may succumb to photobleaching, making them unsuitable challenges in the field, so as to bring PDT to the forefront of
for long-term imaging/activation applications. Recently, codop- oncological diagnosis and intervention. The future of PDT lies
ing with a second sensitizer such as neodymium (Nd3+) (that in the development of a single versatile and efficient
has a absorption maxima around 800 nm) was shown to absorb nanoparticle that encompasses its applicability in both
NIR light at 800 nm following which it sensitizes Yb3+, which bioimaging as well as PDT.
then initiates a typical upconversion process with the activator
ions.282a,288 Although, this technique seem quite promising in AUTHOR INFORMATION
alleviating the short penetration depth and overheating effect
associated 980 nm excitation, no in vitro or in vivo work using Corresponding Author
such UCNs in PDT has been reported until date. *E-mail: biezy@nus.edu.sg.
PDT using UCN is definitely a promising treatment Notes
modality; however, currently it faces many limitations apart
from the issues pertaining to the UCN itself. Optimal PS The authors declare no competing financial interest.
loading is one of the critical factors that determine the Biographies
efficiency of this modality. Currently, majority of the in vivo
studies demonstrated administration of the nanoconstructs via
intratumoral route. This was partly due to premature leakage of
the PS resulting in reduction in the amount of PS ultimately
reaching the target site, and partly due to the lack of specificity
of the nanoconstructs to the tumor tissue and accumulation in
other organs leading to acute toxicity. Although several studies
reported modification of the UCN surface with biocompatible
materials and addition of tumor homing motifs for selective
accumulation of the nanoparticles to the tumor, the percentage
of the injected dose that reached the tumor following
intravenous injection was relatively low, resulting in sub-
optimal PDT and tumor recurrence in a majority of the studies.
This indicates the importance of optimizing the molecular
weight, surface structural conformation, and surface coverage
ratio of the biocompatible polymer like PEG that is grafted to Sasidharan S. Lucky graduated with a Master’s degree in
improve stealth properties of UCNs.289 Furthermore, it is well- Biotechnology in 2004. Thereafter, she worked in Tan Chin Tuan
known that the presence of targeting ligands at high densities Laboratory of Optical Imaging and Photodynamic Therapy for over 4
could promote nonspecific interactions with endothelial and years, before embarking on a Ph.D. under the guidance of Prof. Yong
other noncancerous cells and trigger immune response Zhang and Prof. Khee Chee Soo at the NUS Graduate School of
resulting in opsonization-mediated clearance of nanoparticles Integrative Science and Engineering, National University of Singapore.
from the circulation.290 Development of truly tumor “homing” Her research focuses on the application of upconversion nanoparticles
nanoparticles might require stringent optimization of the for photodynamic therapy of cancer.
number of targeting ligands on the surface of nanoparticles,
targeting multiple receptors that are overexpressed in the tumor
as well as tumor vasculature, and inclusion of cleavable
“antibiofouling agents” that mask the targeting ligands until it
reaches the tumor. At the same time, it must be noted that
most of the in vivo toxicology profiling is limited to small
rodents like mice, which may not be translatable to the clinics.
Moreover, often the lengths of such studies that evaluate the
therapeutic and toxicological effects are relatively short (2 week
and 3 months respectively). As UCNs are generally unable to
degrade into biologically benign components, there exists a lot
of uncertainties in terms of their long-term therapeutic effects,
systemic toxicity as well as clearance. Extensive PK−PD
analysis, together with long-term toxicity and clearance studies,
remains unexplored and deserves immediate attention. It is
clear that UCNs have the potential of being developed into a Khee Chee Soo obtained his medical degree in 1975 from University
multimodality theranostic probe, so that it can be simulta- of Singapore’s Medical School and has many years of experience in the
neously used both as a multimodality imaging tool as well as a clinical application of PDT for treatment of head and neck cancers. He
therapeutic agent. Nevertheless, there is still much room for is the Director of National Cancer Centre Singapore, Professor of
improvement in terms of reduction in size of the construct, Surgery at Yong Loo Lin School of Medicine and Vice Dean of
enhancing the upconversion efficiency, stability in PS loading, Clinical and Faculty Affairs at Duke-NUS Graduate Medical School
optimization of PDT parameters, and thereby increasing the Singapore. He continues to champion the use of PDT as an alternative
PDT efficacy. Owing to the multidisciplinary nature of this cancer treatment option by actively recruiting head and neck cancer

AU DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

patients for pilot clinical trials of fluorescence diagnostic imaging and (10) Konan, Y. N.; Gurny, R.; Allemann, E. J. Photochem. Photobiol., B
photodynamic therapy. 2002, 66, 89.
(11) Castano, A. P.; Demidova, T. N.; Hamblin, M. R. Photodiagn.
Photodyn. Ther. 2004, 1, 279.
(12) Foote, C. S. In Mechanisms of photo-oxygenation; Alan R. Liss:
New York, 1984.
(13) Macdonald, I. J.; Dougherty, T. J. J. Porphyrins Phthalocyanines
2001, 05, 105.
(14) (a) Pineiro, M.; Pereira, M. M.; Formosinho, S. J.; Arnaut, L. G.
J. Phys. Chem. A 2002, 106, 3787. (b) Ding, H.; Yu, H.; Dong, Y.; Tian,
R.; Huang, G.; Boothman, D. A.; Sumer, B. D.; Gao, J. J. Controlled
Release 2011, 156, 276.
(15) (a) Henderson, B. W.; Dougherty, T. J. Photochem. Photobiol.
1992, 55, 145. (b) Dolmans, D. E.; Fukumura, D.; Jain, R. K. Nat. Rev.
Cancer 2003, 3, 380. (c) Dougherty, T. J.; Gomer, C. J.; Henderson, B.
W.; Jori, G.; Kessel, D.; Korbelik, M.; Moan, J.; Peng, Q. J. Natl. Cancer
Inst. 1998, 90, 889.
(16) Niedre, M.; Patterson, M. S.; Wilson, B. C. Photochem. Photobiol.
Yong Zhang is a Professor at Department of Biomedical Engineering, 2002, 75, 382.
National University of Singapore (NUS), a senior Faculty member of (17) Moan, J.; Berg, K. Photochem. Photobiol. 1991, 53, 549.
NUS Nanoscience & Nanotechnology Initiative (NUSNNI/Nano- (18) (a) Mroz, P.; Yaroslavsky, A.; Kharkwal, G. B.; Hamblin, M. R.
Core) and NUS Graduate School for Integrative Sciences and Cancers 2011, 3, 2516. (b) Buytaert, E.; Dewaele, M.; Agostinis, P.
Engineering (NGS). His current research interests include nano- Biochim. Biophys. Acta 2007, 1776, 86.
biophotonics, nanomedicine, biomedical microdevices, and tissue (19) Baluk, P.; Hashizume, H.; McDonald, D. M. Curr. Opin. Genet.
engineering. Dev. 2005, 15, 102.
(20) Chen, B.; Pogue, B. W.; Hoopes, P. J.; Hasan, T. Crit. Rev.
Eukaryot. Gene Expr. 2006, 16, 279.
ACKNOWLEDGMENTS (21) Huang, Z.; Xu, H.; Meyers, A. D.; Musani, A. I.; Wang, L.; Tagg,
The authors would like to acknowledge the funding support R.; Barqawi, A. B.; Chen, Y. K. Technol. Cancer Res. Treat. 2008, 7, 309.
(22) Dolmans, D. E.; Kadambi, A.; Hill, J. S.; Waters, C. A.;
from National Medical Research Council (NMRC) CBRG
Robinson, B. C.; Walker, J. P.; Fukumura, D.; Jain, R. K. Cancer Res.
grant (CBRG13nov052) and Agency for Science, Technology 2002, 62, 2151.
and Research (A*STAR) Biomedical Engineering Programme: (23) Abels, C. Photochem. Photobiol. Sci. 2004, 3, 765.
Grants R-397-000-128-305 (NUS) and BMRNI11133 (24) (a) van Duijnhoven, F. H.; Aalbers, R. I.; Rovers, J. P.; Terpstra,
(NCCS). S.S.L. is a recipient of NGS scholarship from NUS O. T.; Kuppen, P. J. Immunobiology 2003, 207, 105. (b) Nowis, D.;
graduate school (NGS) for Integrative Sciences and Engineer- Stokłosa, T.; Legat, M.; Issat, T.; Jakóbisiak, M.; Gołąb, J. Photodiagn.
ing, National University of Singapore. Photodyn. Ther. 2005, 2, 283.
(25) Allison, R. R.; Moghissi, K. Clin. Endosc. 2013, 46, 24.
REFERENCES (26) Allison, R. R.; Downie, G. H.; Cuenca, R.; Hu, X. H.; Childs, C.
J.; Sibata, C. H. Photodiagn. Photodyn. Ther. 2004, 1, 27.
(1) Ferlay, J.; Shin, H. R.; Bray, F.; Forman, D.; Mathers, C.; Parkin, (27) Ormond, A.; Freeman, H. Materials 2013, 6, 817.
D. M.; International Agency for Research on Cancer; Lyon, France, (28) Josefsen, L. B.; Boyle, R. W. Met.-Based Drugs 2008, 2008,
2008. 276109.
(2) Dougherty, T. J.; Grindey, G. B.; Fiel, R.; Weishaupt, K. R.; (29) Peng, Q.; Moan, J.; Nesland, J. M. Ultrastruct. Pathol. 1996, 20,
Boyle, D. G. J. Natl. Cancer Inst. 1975, 55, 115. 109.
(3) (a) Oleinick, N. L.; Morris, R. L.; Belichenko, I. Photochem. (30) Castano, A. P.; Demidova, T. N.; Hamblin, M. R. Photodiagn.
Photobiol. Sci. 2002, 1, 1. (b) Sharman, W. M.; Allen, C. M.; van Lier, J. Photodyn. Ther. 2005, 2, 91.
E. Methods Enzymol. 2000, 319, 376. (31) Rück, A.; Steiner, R. Minim. Invasive Ther. Allied Technol. 1998,
(4) Yavari, N.; Andersson-Engels, S.; Segersten, U.; Malmstrom, P. U.
7, 503.
Can. J. Urol. 2011, 18, 5778. (32) (a) Konan, Y. N.; Gurny, R.; Allémann, E. J. Photochem.
(5) (a) Allison, R.; Moghissi, K.; Downie, G.; Dixon, K. Photodiagn.
Photobiol. B: Biol. 2002, 66, 89. (b) Lim, C.-K.; Heo, J.; Shin, S.; Jeong,
Photodyn. Ther. 2011, 8, 231. (b) Moghissi, K.; Dixon, K. Photodiagn.
Photodyn. Ther. 2008, 5, 10. K.; Seo, Y. H.; Jang, W.-D.; Park, C. R.; Park, S. Y.; Kim, S.; Kwon, I.
(6) Hur, C.; Nishioka, N. S.; Gazelle, G. S. Dig. Dis. Sci. 2003, 48, C. Cancer Lett. 2012.
1273. (33) Boyle, R. W.; Dolphin, D. Photochem. Photobiol. 1996, 64, 469.
(7) Green, B.; Cobb, A. R. M.; Hopper, C. Br. J. Oral Maxillofac. Surg. (34) Patito, I. A.; Rothmann, C.; Malik, Z. Biol. Cell. 2001, 93, 285.
2013, 51, 283. (35) Dummin, H.; Cernay, T.; Zimmermann, H. W. J. Photochem.
(8) Kostovic, K.; Pastar, Z.; Ceovic, R.; Mokos, Z. B.; Buzina, D. S.; Photobiol., B 1997, 37, 219.
Stanimirovic, A. Coll. Antropol. 2012, 36, 1477. (36) (a) Hudson, R.; Carcenac, M.; Smith, K.; Madden, L.; Clarke,
(9) (a) Muragaki, Y.; Akimoto, J.; Maruyama, T.; Iseki, H.; Ikuta, S.; O. J.; Pelegrin, A.; Greenman, J.; Boyle, R. W. Br. J. Cancer 2005, 92,
Nitta, M.; Maebayashi, K.; Saito, T.; Okada, Y.; Kaneko, S.; 1442. (b) Malatesti, N.; Smith, K.; Savoie, H.; Greenman, J.; Boyle, R.
Matsumura, A.; Kuroiwa, T.; Karasawa, K.; Nakazato, Y.; Kayama, T. W. Int. J. Oncol. 2006, 28, 1561. (c) Staneloudi, C.; Smith, K. A.;
J. Neurosurg. 2013, 119, 845. (b) Rigual, N. R.; et al. JAMA Otolaryngol. Hudson, R.; Malatesti, N.; Savoie, H.; Boyle, R. W.; Greenman. J.
Head Neck Surg. 2013, 139, 706. (c) Friedberg, J. S.; Mick, R.; Immunol. 2007, 120, 512.
Stevenson, J. P.; Zhu, T.; Busch, T. M.; Shin, D.; Smith, D.; Culligan, (37) Taquet, J. P.; Frochot, C.; Manneville, V.; Barberi-Heyob, M.
M.; Dimofte, A.; Glatstein, E.; Hahn, S. M. J. Clin. Oncol. 2004, 22, Curr. Med. Chem. 2007, 14, 1673.
2192. (d) Akimoto, J.; Haraoka, J.; Aizawa, K. Photodiagn. Photodyn. (38) Allison, R. R. Future Oncol. 2014, 10, 123.
Ther. 2012, 9, 91. (39) Zhu, T. C.; Finlay, J. C. Med. Phys. 2008, 35, 3127.

AV DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

(40) (a) Wilson, B. C.; Patterson, M. S.; Lilge, L. Laser Med. Sci. (66) Bechet, D.; Couleaud, P.; Frochot, C.; Viriot, M. L.; Guillemin,
1997, 12, 182. (b) Finlay, J. C.; Conover, D. L.; Hull, E. L.; Foster, T. F.; Barberi-Heyob, M. Trends Biotechnol. 2008, 26, 612.
H. Photochem. Photobiol. 2001, 73, 54. (67) (a) Paszko, E.; Ehrhardt, C.; Senge, M. O.; Kelleher, D. P.;
(41) Jarvi, M. T.; Niedre, M. J.; Patterson, M. S.; Wilson, B. C. Reynolds, J. V. Photodiagn. Photodyn. Ther. 2011, 8, 14. (b) Chatterjee,
Photochem. Photobiol. 2006, 82, 1198. D. K.; Fong, L. S.; Zhang, Y. Adv. Drug Delivery Rev. 2008, 60, 1627.
(42) Fingar, V. H.; Wieman, T. J.; Park, Y. J.; Henderson, B. W. J. (68) Birrenbach, G.; Speiser, P. P. J. Pharm. Sci. 1976, 65, 1763.
Surg. Res. 1992, 53, 524. (69) Labib, A.; Lenaerts, V.; Chouinard, F.; Leroux, J.-C.; Ouellet, R.;
(43) (a) Puizina-Ivic, N.; Zorc, H.; Vanjaka-Rogosic, L.; Miric, L.; van Lier, J. Pharm. Res. 1991, 8, 1027.
Persin, A. Coll. Antropol. 2008, 32 (Suppl 2), 67. (b) Ascencio, M.; (70) (a) Allemann, E.; Brasseur, N.; Benrezzak, O.; Rousseau, J.;
Estevez, J. P.; Delemer, M.; Farine, M. O.; Collinet, P.; Mordon, S. Kudrevich, S. V.; Boyle, R. W.; Leroux, J. C.; Gurny, R.; Van Lier, J. E.
Photodiagn. Photodyn. Ther. 2008, 5, 210. (c) Pogue, B. W.; Hasan, T. J. Pharm. Pharmacol. 1995, 47, 382. (b) Konan, Y. N.; Berton, M.;
Radiat. Res. 1997, 147, 551. Gurny, R.; Allemann, E. Eur. J. Pharm. Sci. 2003, 18, 241. (c) Allemann,
(44) (a) Henderson, B. W.; Busch, T. M.; Vaughan, L. A.; Frawley, E.; Rousseau, J.; Brasseur, N.; Kudrevich, S. V.; Lewis, K.; van Lier, J.
N. P.; Babich, D.; Sosa, T. A.; Zollo, J. D.; Dee, A. S.; Cooper, M. T.; E. Int. J. Cancer 1996, 66, 821. (d) Konan, Y. N.; Chevallier, J.; Gurny,
Bellnier, D. A.; Greco, W. R.; Oseroff, A. R. Cancer Res. 2000, 60, 525. R.; Allémann, E. Photochem. Photobiol. 2003, 77, 638. (e) McCarthy, J.
(b) Finlay, J. C.; Conover, D. L.; Hull, E. L.; Foster, T. H. Photochem. R.; Perez, J. M.; Bruckner, C.; Weissleder, R. Nano Lett. 2005, 5, 2552.
Photobiol. 2001, 73, 54. (c) Middelburg, T. A.; Van Zaane, F.; De (71) (a) da Silva, C. L.; Del Ciampo, J. O.; Rossetti, F. C.; Bentley,
Bruijn, H. S.; Van Der Ploeg-van den Heuvel, A.; Sterenborg, H. J.; M. V.; Pierre, M. B. J. Nanosci. Nanotechnol. 2013, 13, 6533. (b) Rojnik,
Neumann, H. A.; De Haas, E. R.; Robinson, D. J. Photochem. Photobiol. M.; Kocbek, P.; Moret, F.; Compagnin, C.; Celotti, L.; Bovis, M. J.;
2010, 86, 1140. Woodhams, J. H.; Macrobert, A. J.; Scheglmann, D.; Helfrich, W.;
(45) Chen, Q.; Huang, Z.; Chen, H.; Shapiro, H.; Beckers, J.; Hetzel, Verkaik, M. J.; Papini, E.; Reddi, E.; Kos, J. Nanomedicine (Lond) 2012,
F. W. Photochem. Photobiol. 2002, 76, 197. 7, 663. (c) Fadel, M.; Kassab, K.; Fadeel, D. A. Laser Med. Sci. 2010,
(46) Huang, Z.; Chen, Q.; Shakil, A.; Chen, H.; Beckers, J.; Shapiro, 25, 283. (d) da Silva, A. R.; Inada, N. M.; Rettori, D.; Baratti, M. O.;
H.; Hetzel, F. W. Photochem. Photobiol. 2003, 78, 496. Vercesi, A. E.; Jorge, R. A. J. Photochem. Photobiol., B 2009, 94, 101.
(47) Brahimi-Horn, M. C.; Pouyssegur, J. Int. Rev. Cytol. 2005, 242,
(e) Zeisser-Labouebe, M.; Lange, N.; Gurny, R.; Delie, F. Int. J. Pharm.
157.
2006, 326, 174.
(48) (a) Wang, G. L.; Semenza, G. L. J. Biol. Chem. 1995, 270, 1230.
(72) Lu, J. M.; Wang, X.; Marin-Muller, C.; Wang, H.; Lin, P. H.;
(b) Wang, G. L.; Jiang, B. H.; Rue, E. A.; Semenza, G. L. Proc. Natl.
Yao, Q.; Chen, C. Expert Rev. Mol. Diagn. 2009, 9, 325.
Acad. Sci. U.S.A. 1995, 92, 5510.
(73) Lee, Y.-E.; Kopelman, R. In Biomedical Nanotechnology; Hurst, S.
(49) Ratcliffe, P. J.; O’Rourke, J. F.; Maxwell, P. H.; Pugh, C. W. J.
J., Ed.; Humana Press: New York, 2011; Vol. 726.
Exp. Biol. 1998, 201, 1153.
(74) (a) Shive, M. S.; Anderson, J. M. Adv. Drug Delivery Rev. 1997,
(50) (a) Bhuvaneswari, R.; Gan, Y.; Soo, K.; Olivo, M. Cell. Mol. Life
Sci. 2009, 66, 2275. (b) Bhuvaneswari, R.; Gan, Y. Y.; Lucky, S. S.; 28, 5. (b) Schakenraad, J. M.; Hardonk, M. J.; Feijen, J.; Molenaar, I.;
Chin, W. W.; Ali, S. M.; Soo, K. C.; Olivo, M. Mol. Cancer 2008, 7, 56. Nieuwenhuis, P. J. Biomed. Mater. Res. 1990, 24, 529.
(c) Ferrario, A.; von Tiehl, K. F.; Rucker, N.; Schwarz, M. A.; Gill, P. (75) (a) Avgoustakis, K.; Beletsi, A.; Panagi, Z.; Klepetsanis, P.;
S.; Gomer, C. J. Cancer Res. 2000, 60, 4066. (d) Zhou, Q.; Olivo, M.; Livaniou, E.; Evangelatos, G.; Ithakissios, D. S. Int. J. Pharm. 2003,
Lye, K. Y.; Moore, S.; Sharma, A.; Chowbay, B. Cancer Chemother. 259, 115. (b) Beletsi, A.; Panagi, Z.; Avgoustakis, K. Int. J. Pharm.
Pharmacol. 2005, 56, 569. 2005, 298, 233.
(51) (a) Allison, R. R.; Bagnato, V. S.; Sibata, C. H. Future Oncol. (76) Panagi, Z.; Beletsi, A.; Evangelatos, G.; Livaniou, E.; Ithakissios,
2010, 6, 929. (b) Kudinova, N. V.; Berezov, T. T. Biochem. Moscow D. S.; Avgoustakis, K. Int. J. Pharm. 2001, 221, 143.
Suppl. Ser. B 2010, 4, 95. (77) Master, A. M.; Qi, Y.; Oleinick, N. L.; Gupta, A. S. Nanomedicine
(52) Ricchelli, F. J. Photochem. Photobiol., B 1995, 29, 109. 2012, 8, 655.
(53) Taillefer, J.; Jones, M. C.; Brasseur, N.; van Lier, J. E.; Leroux, J. (78) Peng, C. L.; Yang, L. Y.; Luo, T. Y.; Lai, P. S.; Yang, S. J.; Lin, W.
C. J. Pharm. Sci. 2000, 89, 52. J.; Shieh, M. J. Nanotechnology 2010, 21, 155103.
(54) Frangioni, J. V. Curr. Opin. Chem. Biol. 2003, 7, 626. (79) Kim, W. L.; Cho, H.; Li, L.; Kang, H. C.; Huh, K. M.
(55) Oleinick, N. L.; Evans, H. H. Radiat. Res. 1998, 150, S146. Biomacromolecules 2014, 15, 2224.
(56) Ronald, W. W.; Marwood, N. E. In Electro-Optics Handbook; (80) Gupta, A.; Wang, S.; Pera, P.; Rao, K. V.; Patel, N.;
2nd ed.; McGraw Hill: New York, 2000; Professional Access Ohulchanskyy, T. Y.; Missert, J.; Morgan, J.; Koo-Lee, Y. E.;
Engineering. Kopelman, R.; Pandey, R. K. Nanomedicine 2012, 8, 941.
(57) (a) Lu, T.; Shao, P.; Mathew, I.; Sand, A.; Sun, W. J. Am. Chem. (81) Lee, D. J.; Park, G. Y.; Oh, K. T.; Oh, N. M.; Kwag, D. S.; Youn,
Soc. 2008, 130, 15782. (b) Luan, L.; Ding, L.; Zhang, W.; Shi, J.; Yu, Y. S.; Oh, Y. T.; Park, J. w.; Lee, E. S. Int. J. Pharm. 2012, 434, 257.
X.; Liu, W. Bioorg. Med. Chem. Lett. 2013, 23, 3775. (82) Wang, S.; Kim, G.; Lee, Y.-E. K.; Hah, H. J.; Ethirajan, M.;
(58) Honzak, L.; Sentjurc, M.; Swartz, H. M. J. Controlled Release Pandey, R. K.; Kopelman, R. ACS Nano 2012, 6, 6843.
2000, 66, 221. (83) Yoon, H.-J.; Lim, T. G.; Kim, J.-H.; Cho, Y. M.; Kim, Y. S.;
(59) (a) Prasad, P. N. Curr. Opin. Solid State Mater. Sci. 2004, 8, 11. Chung, U. S.; Kim, J. H.; Choi, B. W.; Koh, W.-G.; Jang, W.-D.
(b) Jain, K. K. Technol. Cancer Res. Treat. 2008, 7, 1. (c) Allison, R. R.; Biomacromolecules 2014, 15, 1382.
Mota, H. C.; Bagnato, V. S.; Sibata, C. H. Photodiagn. Photodyn. Ther. (84) Xiang, G. H.; Hong, G. B.; Wang, Y.; Cheng, D.; Zhou, J. X.;
2008, 5, 19. Shuai, X. T. Int. J. Nanomed. 2013, 8, 4613.
(60) Davis, M. E.; Chen, Z.; Shin, D. M. Nat. Rev. Drug Discovery (85) Gong, H.; Dong, Z.; Liu, Y.; Yin, S.; Cheng, L.; Xi, W.; Xiang, J.;
2008, 7, 771. Liu, K.; Li, Y.; Liu, Z. Adv. Funct. Mater. 2014, 24, 6492.
(61) Konan-Kouakou, Y. N.; Boch, R.; Gurny, R.; Allémann, E. J. (86) Bangham, A. D. Chem. Phys. Lipids 1993, 64, 275.
Controlled Release 2005, 103, 83. (87) Torchilin, V. P. Nat. Rev. Drug Discovery 2005, 4, 145.
(62) Master, A.; Livingston, M.; Sen Gupta, A. J. Controlled Release (88) Vemuri, S.; Rhodes, C. T. Pharm. Acta Helv. 1995, 70, 95.
2013, 168, 88. (89) Derycke, A. S.; de Witte, P. A. Adv. Drug Delivery Rev. 2004, 56,
(63) Allison, R. R.; Bagnato, V. S.; Sibata, C. H. Future Oncol. 2010, 17.
6, 929. (90) Lasic, D. D.; Martin, F. J.; Gabizon, A.; Huang, S. K.;
(64) Maeda, H.; Wu, J.; Sawa, T.; Matsumura, Y.; Hori, K. J. Papahadjopoulos, D. Biochim. Biophys. Acta 1991, 1070, 187.
Controlled Release 2000, 65, 271. (91) Chowdhary, R. K.; Shariff, I.; Dolphin, D. J. Pharm. Pharm. Sci.
(65) Reddi, E. J. Photochem. Photobiol. B: Biol. 1997, 37, 189. 2003, 6, 13.

AW DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

(92) (a) Ichikawa, K.; Hikita, T.; Maeda, N.; Takeuchi, Y.; Namba, Martins, O. P.; Simioni, A. R.; Morais, P. C.; Azevedo, R. B.; Tedesco,
Y.; Oku, N. Biol. Pharm. Bull. 2004, 27, 443. (b) Sadzuka, Y.; Iwasaki, A. C.; Lacava, Z. G. J. Nanobiotechnol. 2013, 11, 41.
F.; Sugiyama, I.; Horiuchi, K.; Hirano, T.; Ozawa, H.; Kanayama, N.; (118) Hejazi, R.; Amiji, M. J. Controlled Release 2003, 89, 151.
Oku, N. Toxicol. Lett. 2008, 182, 110. (119) (a) Cho, Y. W.; Park, S. A.; Han, T. H.; Son, D. H.; Park, J. S.;
(93) Rahmanzadeh, R.; Rai, P.; Celli, J. P.; Rizvi, I.; Baron-Luhr, B.; Oh, S. J.; Moon, D. H.; Cho, K. J.; Ahn, C. H.; Byun, Y.; Kim, I. S.;
Gerdes, J.; Hasan, T. Cancer Res. 2010, 70, 9234. Kwon, I. C.; Kim, S. Y. Biomaterials 2007, 28, 1236. (b) Kim, J. H.;
(94) Oku, N.; Ishii, T. Methods Enzymol. 2009, 465, 313. Kim, Y. S.; Park, K.; Kang, E.; Lee, S.; Nam, H. Y.; Kim, K.; Park, J. H.;
(95) Moret, F.; Scheglmann, D.; Reddi, E. Photochem. Photobiol. Sci. Chi, D. Y.; Park, R. W.; Kim, I. S.; Choi, K.; Chan Kwon, I.
2013, 12, 823. Biomaterials 2008, 29, 1920. (c) Kim, J. H.; Kim, Y. S.; Kim, S.; Park, J.
(96) (a) Paszko, E.; Vaz, G. M.; Ehrhardt, C.; Senge, M. O. Eur. J. H.; Kim, K.; Choi, K.; Chung, H.; Jeong, S. Y.; Park, R. W.; Kim, I. S.;
Pharm. Sci. 2013, 48, 202. (b) Derycke, A. S.; Kamuhabwa, A.; Gijsens, Kwon, I. C. J. Controlled Release 2006, 111, 228.
A.; Roskams, T.; De Vos, D.; Kasran, A.; Huwyler, J.; Missiaen, L.; de (120) (a) Lee, S. J.; Park, K.; Oh, Y. K.; Kwon, S. H.; Her, S.; Kim, I.
Witte, P. A. J. Natl. Cancer Inst. 2004, 96, 1620. S.; Choi, K.; Kim, H.; Lee, S. G.; Kim, K.; Kwon, I. C. Biomaterials
(97) Jin, C. S.; Zheng, G. Lasers Surg. Med. 2011, 43, 734. 2009, 30, 2929. (b) Chung, C. W.; Chung, K. D.; Jeong, Y. I.; Kang, D.
(98) Skupin-Mrugalska, P.; Piskorz, J.; Goslinski, T.; Mielcarek, J.; H. Int. J. Nanomed. 2013, 8, 809.
Konopka, K.; Düzgüneş, N. Drug Discovery Today 2013, 18, 776. (121) (a) Lee, S. J.; Koo, H.; Lee, D.-E.; Min, S.; Lee, S.; Chen, X.;
(99) (a) Simoes, S.; Moreira, J. N.; Fonseca, C.; Duzgunes, N.; de Choi, Y.; Leary, J. F.; Park, K.; Jeong, S. Y.; Kwon, I. C.; Kim, K.; Choi,
Lima, M. C. Adv. Drug Delivery Rev. 2004, 56, 947. (b) Yavlovich, A.; K. Biomaterials 2011, 32, 4021. (b) Oh, I.-h.; Min, H. S.; Li, L.; Tran,
Smith, B.; Gupta, K.; Blumenthal, R.; Puri, A. Mol. Membr. Biol. 2010, T. H.; Lee, Y.-k.; Kwon, I. C.; Choi, K.; Kim, K.; Huh, K. M.
27, 364. Biomaterials 2013, 34, 6454.
(100) Reshetov, V.; Lassalle, H. P.; Francois, A.; Dumas, D.; Hupont, (122) Lee, H. M.; Jeong, Y.-I.; Kim, D. H.; Kwak, T. W.; Chung, C.-
S.; Grafe, S.; Filipe, V.; Jiskoot, W.; Guillemin, F.; Zorin, V.; W.; Kim, C. H.; Kang, D. H. Int. J. Pharm. 2013, 454, 74.
Bezdetnaya, L. Int. J. Nanomed. 2013, 8, 3817. (123) Chen, R.; Wang, X.; Yao, X.; Zheng, X.; Wang, J.; Jiang, X.
(101) Spring, B.; Mai, Z.; Rai, P.; Chang, S.; Hasan, T. Proc. SPIE Biomaterials 2013, 34, 8314.
2010, 7551, 755104. (124) Kommareddy, S.; Amiji, M. Nanomedicine 2007, 3, 32.
(102) Lovell, J. F.; Jin, C. S.; Huynh, E.; Jin, H.; Kim, C.; Rubinstein, (125) Babu, A.; Jeyasubramanian, K.; Gunasekaran, P.; Murugesan, R.
J. L.; Chan, W. C.; Cao, W.; Wang, L. V.; Zheng, G. Nat. Mater. 2011, J. Biomed. Nanotechnol. 2012, 8, 43.
10, 324. (126) Babu, A.; Periasamy, J.; Gunasekaran, A.; Kumaresan, G.;
(103) Liang, X.; Li, X.; Yue, X.; Dai, Z. Angew. Chem., Int. Ed. 2011, Naicker, S.; Gunasekaran, P.; Murugesan, R. J. Biomed. Nanotechnol
50, 11622. 2013, 9, 177.
(104) Mir, Y.; Elrington, S. A.; Hasan, T. Nanomedicine 2013, 9, (127) Stöber, W.; Fink, A.; Bohn, E. J. Colloid Interface Sci. 1968, 26,
1114. 62.
(105) Svenson, S.; Tomalia, D. A. Adv. Drug Delivery Rev. 2005, 57, (128) Das, S.; Jain, T. K.; Maitra, A. J. Colloid Interface Sci. 2002, 252,
2106. 82.
(106) (a) Boas, U.; Heegaard, P. M. H. Chem. Soc. Rev. 2004, 33, 43. (129) Couleaud, P.; Morosini, V.; Frochot, C.; Richeter, S.; Raehm,
(b) Wolinsky, J. B.; Grinstaff, M. W. Adv. Drug Delivery Rev. 2008, 60, L.; Durand, J.-O. Nanoscale 2010, 2, 1083.
1037. (130) Roy, I.; Ohulchanskyy, T. Y.; Pudavar, H. E.; Bergey, E. J.;
(107) Klajnert, B.; Rozanek, M.; Bryszewska, M. Curr. Med. Chem. Oseroff, A. R.; Morgan, J.; Dougherty, T. J.; Prasad, P. N. J. Am. Chem.
2012, 19, 4903. Soc. 2003, 125, 7860.
(108) Hackbarth, S.; Horneffer, V.; Wiehe, A.; Hillenkamp, F.; Röder, (131) Ohulchanskyy, T. Y.; Roy, I.; Goswami, L. N.; Chen, Y.;
B. Chem. Phys. 2001, 269, 339. Bergey, E. J.; Pandey, R. K.; Oseroff, A. R.; Prasad, P. N. Nano Lett.
(109) Battah, S. H.; Chee, C. E.; Nakanishi, H.; Gerscher, S.; 2007, 7, 2835.
MacRobert, A. J.; Edwards, C. Bioconjugate Chem. 2001, 12, 980. (132) Zhao, B.; Yin, J. J.; Bilski, P. J.; Chignell, C. F.; Roberts, J. E.;
(110) (a) Battah, S.; O’Neill, S.; Edwards, C.; Balaratnam, S.; Dobbin, He, Y. Y. Toxicol. Appl. Pharmacol. 2009, 241, 163.
P.; MacRobert, A. J. Int. J. Biochem. Cell Biol. 2006, 38, 1382. (b) Di (133) Simon, V.; Devaux, C.; Darmon, A.; Donnet, T.; Thienot, E.;
Venosa, G. M.; Casas, A. G.; Battah, S.; Dobbin, P.; Fukuda, H.; Germain, M.; Honnorat, J.; Duval, A.; Pottier, A.; Borghi, E.; Levy, L.;
MacRobert, A. J.; Batlle, A. Int. J. Biochem. Cell Biol. 2006, 38, 82. Marill, J. Photochem. Photobiol. 2010, 86, 213.
(c) Battah, S.; Balaratnam, S.; Casas, A.; O’Neill, S.; Edwards, C.; (134) (a) Selvestrel, F.; Moret, F.; Segat, D.; Woodhams, J. H.;
Batlle, A.; Dobbin, P.; MacRobert, A. J. Mol. Cancer Ther. 2007, 6, 876. Fracasso, G.; Echevarria, I. M.; Bau, L.; Rastrelli, F.; Compagnin, C.;
(d) Casas, A.; Battah, S.; Di Venosa, G.; Dobbin, P.; Rodriguez, L.; Reddi, E.; Fedeli, C.; Papini, E.; Tavano, R.; MacKenzie, A.; Bovis, M.;
Fukuda, H.; Batlle, A.; MacRobert, A. J. J. Controlled Release 2009, 135, Yaghini, E.; MacRobert, A. J.; Zanini, S.; Boscaini, A.; Colombatti, M.;
136. Mancin, F. Nanoscale 2013, 5, 6106. (b) Compagnin, C.; Bau, L.;
(111) (a) Nishiyama, N.; Stapert, H. R.; Zhang, G.-D.; Takasu, D.; Mognato, M.; Celotti, L.; Miotto, G.; Arduini, M.; Moret, F.; Fede, C.;
Jiang, D.-L.; Nagano, T.; Aida, T.; Kataoka, K. Bioconj. Chem. 2002, 14, Selvestrel, F.; Rio Echevarria, I. M.; Mancin, F.; Reddi, E.
58. (b) Zhang, G.-D.; Nishiyama, N.; Harada, A.; Jiang, D.-L.; Aida, T.; Nanotechnology 2009, 20, 345101.
Kataoka, K. Macromolecules 2003, 36, 1304. (c) Zhang, G.-D.; Harada, (135) Tang, W.; Xu, H.; Kopelman, R.; Philbert, M. A. Photochem.
A.; Nishiyama, N.; Jiang, D.-L.; Koyama, H.; Aida, T.; Kataoka, K. J. Photobiol. 2005, 81, 242.
Controlled Release 2003, 93, 141. (136) Qian, J.; Gharibi, A.; He, S. J. Biomed. Opt. 2009, 14, 014012.
(112) Taratula, O.; Schumann, C.; Naleway, M. A.; Pang, A. J.; Chon, (137) Brevet, D.; Gary-Bobo, M.; Raehm, L.; Richeter, S.; Hocine,
K. J.; Taratula, O. Mol. Pharmaceutics 2013, 10, 3946. O.; Amro, K.; Loock, B.; Couleaud, P.; Frochot, C.; Morere, A.;
(113) Gradishar, W. J. Exp. Opin. Pharmacother. 2006, 7, 1041. Maillard, P.; Garcia, M.; Durand, J.-O. Chem. Commun. 2009, 1475.
(114) Wacker, M.; Chen, K.; Preuss, A.; Possemeyer, K.; Roeder, B.; (138) Zhang, R.; Wu, C.; Tong, L.; Tang, B.; Xu, Q.-H. Langmuir
Langer, K. Int. J. Pharm. 2010, 393, 254. 2009, 25, 10153.
(115) Kratz, F. J. Controlled Release 2008, 132, 171. (139) Teng, I. T.; Chang, Y.-J.; Wang, L.-S.; Lu, H.-Y.; Wu, L.-C.;
(116) Jeong, H.; Huh, M.; Lee, S. J.; Koo, H.; Kwon, I. C.; Jeong, S. Yang, C.-M.; Chiu, C.-C.; Yang, C.-H.; Hsu, S.-L.; Ho, J.-a. A.
Y.; Kim, K. Theranostics 2011, 1, 230. Biomaterials 2013, 34, 7462.
(117) Portilho, F. A.; Cavalcanti, C. E.; Miranda-Vilela, A. L.; (140) Zhao, Z. X.; Huang, Y. Z.; Shi, S. G.; Tang, S. H.; Li, D. H.;
Estevanato, L. L.; Longo, J. P.; Almeida Santos Mde, F.; Bocca, A. L.; Chen, X. L. Nanotechnology 2014, 25, 285701.

AX DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

(141) Yang, G.; Gong, H.; Qian, X.; Tan, P.; Li, Z.; Liu, T.; Liu, J.; Li, 2008, 6, 1015. (b) Metanawin, T.; Tang, T.; Chen, R.; Vernon, D.;
Y.; Liu, Z. Nano Res. 2014, 1. Wang, X. Nanotechnology 2011, 22, 235604.
(142) Hone, D. C.; Walker, P. I.; Evans-Gowing, R.; FitzGerald, S.; (168) Kwag, D. S.; Oh, N. M.; Oh, Y. T.; Oh, K. T.; Youn, Y. S.; Lee,
Beeby, A.; Chambrier, I.; Cook, M. J.; Russell, D. A. Langmuir 2002, E. S. Int. J. Pharm. 2012, 431, 204.
18, 2985. (169) Liu, Q.; Xu, L.; Zhang, X.; Li, N.; Zheng, J.; Guan, M.; Fang,
(143) Wieder, M. E.; Hone, D. C.; Cook, M. J.; Handsley, M. M.; X.; Wang, C.; Shu, C. Chem.Asian J. 2013, 8, 2370.
Gavrilovic, J.; Russell, D. A. Photochem. Photobiol. Sci. 2006, 5, 727. (170) Shi, J.; Wang, Z.; Wang, L.; Wang, H.; Li, L.; Yu, X.; Zhang, J.;
(144) Camerin, M.; Magaraggia, M.; Soncin, M.; Jori, G.; Moreno, Ma, R.; Zhang, Z. Int. J. Nanomedicine 2013, 8, 1551.
M.; Chambrier, I.; Cook, M. J.; Russell, D. A. Eur. J. Cancer 2010, 46, (171) Fujishima, A.; Honda, K. Nature 1972, 238, 37.
1910. (172) (a) Rozhkova, E. A.; Ulasov, I.; Lai, B.; Dimitrijevic, N. M.;
(145) Stuchinskaya, T.; Moreno, M.; Cook, M. J.; Edwards, D. R.; Lesniak, M. S.; Rajh, T. Nano Lett. 2009, 9, 3337. (b) Cai, R.;
Russell, D. A. Photochem. Photobiol. Sci. 2011, 10, 822. Hashimoto, K.; Itoh, K.; Kubota, Y.; Fujishima, A. Bull. Chem. Soc. Jpn.
(146) Cheng, Y.; Meyers, J. D.; Broome, A. M.; Kenney, M. E.; 1991, 64, 1268. (c) Cai, R.; Kubota, Y.; Shuin, T.; Sakai, H.;
Basilion, J. P.; Burda, C. J. Am. Chem. Soc. 2011, 133, 2583. Hashimoto, K.; Fujishima, A. Cancer Res. 1992, 52, 2346. (d) Wamer,
(147) Cheng, Y.; Samia, A. C.; Li, J.; Kenney, M. E.; Resnick, A.; W. G.; Yin, J. J.; Wei, R. R. Free Radic. Biol. Med. 1997, 23, 851.
Burda, C. Langmuir 2009, 26, 2248. (173) Murakami, Y.; Kenji, E.; Nosaka, A. Y.; Nosaka, Y. J. Phys.
(148) Jang, B.; Park, J.-Y.; Tung, C.-H.; Kim, I.-H.; Choi, Y. ACS Chem. B 2006, 110, 16808.
Nano 2011, 5, 1086. (174) Nosaka, Y.; Nakamura, M.; Hirakawa, T. Phys. Chem. Chem.
(149) Khlebtsov, B.; Panfilova, E.; Khanadeev, V.; Bibikova, O.; Phys. 2002, 4, 1088.
Terentyuk, G.; Ivanov, A.; Rumyantseva, V.; Shilov, I.; Ryabova, A.; (175) Kubo, W.; Tatsuma, T. Anal. Sci. 2004, 20, 591.
Loshchenov, V.; Khlebtsov, N. G. ACS Nano 2011, 5, 7077. (176) (a) Li, Z.; Mi, L.; Wang, P. N.; Chen, J. Y. Nanoscale Res. Lett.
(150) Gao, L.; Fei, J.; Zhao, J.; Li, H.; Cui, Y.; Li, J. ACS Nano 2012, 2011, 6, 356. (b) Lagopati, N.; Kitsiou, P. V.; Kontos, A. I.; Venieratos,
6, 8030. P.; Kotsopoulou, E.; Kontos, A. G.; Dionysiou, D. D.; Pispas, S.;
(151) Seo, S.-H.; Kim, B.-M.; Joe, A.; Han, H.-W.; Chen, X.; Cheng, Tsilibary, E. C.; Falaras, P. J. Photochem. Photobiol. A: Chem. 2010, 214,
Z.; Jang, E.-S. Biomaterials 2014, 35, 3309. 215.
(152) Cheng, L.; Wang, C.; Feng, L.; Yang, K.; Liu, Z. Chem. Rev. (177) Fujishima, A.; Ohtsuki, J.; Yamashita, T.; Hayakawa, S.
2014. Photomed. Photobiol. 1986, 8, 45.
(153) Wang, S.; Huang, P.; Nie, L.; Xing, R.; Liu, D.; Wang, Z.; Lin, (178) Cai, R.; Hashimoto, K.; Kubota, Y.; Fujishima, A. Chem. Lett.
J.; Chen, S.; Niu, G.; Lu, G.; Chen, X. Adv. Mater. 2013, 25, 3055. 1992, 21, 427.
(154) Basilion, J. P.; Yeon, S.; Botnar, R. In Curr. Top. Dev. Biol.; Eric, (179) Kubota, Y.; Shuin, T.; Kawasaki, C.; Hosaka, M.; Kitamura, H.;
T. A., Ed.; Academic Press: New York, 2005; Vol. 70. Cai, R.; Sakai, H.; Hashimoto, K.; Fujishima, A. Br. J. Cancer 1994, 70,
(155) Kopelman, R.; Lee Koo, Y.-E.; Philbert, M.; Moffat, B. A.; 1107.
Ramachandra Reddy, G.; McConville, P.; Hall, D. E.; Chenevert, T. L.; (180) Huang, N.-p.; Min-hua, X.; Yuan, C.-w.; Rui-rong, Y. J.
Bhojani, M. S.; Buck, S. M.; Rehemtulla, A.; Ross, B. D. J. Magn. Magn. Photochem. Photobiol. A: Chem. 1997, 108, 229.
Mater. 2005, 293, 404. (181) Xu, M.; Huang, N.; Xiao, Z.; Lu, Z. Supramol. Sci. 1998, 5, 449.
(156) Reddy, G. R.; Bhojani, M. S.; McConville, P.; Moody, J.; (182) Zhang, A. P.; Sun, Y. P. World J. Gastroenterol. 2004, 10, 3191.
Moffat, B. A.; Hall, D. E.; Kim, G.; Koo, Y.-E. L.; Woolliscroft, M. J.; (183) Wang, C.; Cao, S.; Tie, X.; Qiu, B.; Wu, A.; Zheng, Z. Mol. Biol.
Sugai, J. V.; Johnson, T. D.; Philbert, M. A.; Kopelman, R.; Rehemtulla, Rep. 2011, 38, 523.
A.; Ross, B. D. Clin. Cancer. Res. 2006, 12, 6677. (184) Jacques, S. L.; Weaver, D. R.; Reppert, S. M. Photochem.
(157) Chen, Z. L.; Sun, Y.; Huang, P.; Yang, X. X.; Zhou, X. P. Photobiol. 1987, 45, 637.
Nanoscale Res. Lett. 2009, 4, 400. (185) Li, J.; Guo, D.; Wang, X.; Wang, H.; Jiang, H.; Chen, B.
(158) Benachour, H.; Seve, A.; Bastogne, T.; Frochot, C.; Nanoscale Res. Lett. 2010, 5, 1063.
Vanderesse, R.; Jasniewski, J.; Miladi, I.; Billotey, C.; Tillement, O.; (186) Hackenberg, S.; Scherzed, A.; Kessler, M.; Froelich, K.;
Lux, F.; Barberi-Heyob, M. Theranostics 2012, 2, 889. Ginzkey, C.; Koehler, C.; Burghartz, M.; Hagen, R.; Kleinsasser, N. Int.
(159) Li, Z.; Wang, C.; Cheng, L.; Gong, H.; Yin, S.; Gong, Q.; Li, Y.; J. Oncol. 2010, 37, 1583.
Liu, Z. Biomaterials 2013, 34, 9160. (187) Hariharan, R.; Senthilkumar, S.; Suganthi, A.; Rajarajan, M. J.
(160) Mikhaylov, G.; Mikac, U.; Magaeva, A. A.; Itin, V. I.; Naiden, E. Photochem. Photobiol. B: Biol. 2012, 116, 56.
P.; Psakhye, I.; Babes, L.; Reinheckel, T.; Peters, C.; Zeiser, R.; Bogyo, (188) Chen, W.; Zhang, J. J. Nanosci. Nanotechnol. 2006, 6, 1159.
M.; Turk, V.; Psakhye, S. G.; Turk, B.; Vasiljeva, O. Nat. Nanotechnol. (189) Chen, W. J. Biomed. Nanotechnol. 2008, 4, 369.
2011, 6, 594. (190) Liu, Y.; Chen, W.; Wang, S.; Joly, A. G. Appl. Phys. Lett. 2008,
(161) Sun, Y.; Chen, Z.-l.; Yang, X.-x.; Huang, P.; Zhou, X.-p.; Du, 92.
X.-x. Nanotechnology 2009, 20, 135102. (191) Zou, X.; Yao, M.; Ma, L.; Hossu, M.; Han, X.; Juzenas, P.;
(162) Huang, P.; Li, Z.; Lin, J.; Yang, D.; Gao, G.; Xu, C.; Bao, L.; Chen, W. Nanomedicine 2014, 1.
Zhang, C.; Wang, K.; Song, H.; Hu, H.; Cui, D. Biomaterials 2011, 32, (192) Eijk, C. W. E. v. Phys. Med. Biol. 2002, 47, R85.
3447. (193) Zhang, J. Z. Acc. Chem. Res. 1997, 30, 423.
(163) Silva, A. K. A.; Kolosnjaj-Tabi, J.; Bonneau, S.; Marangon, I.; (194) Yang, W.; Read, P. W.; Mi, J.; Baisden, J. M.; Reardon, K. A.;
Boggetto, N.; Aubertin, K.; Clément, O.; Bureau, M. F.; Luciani, N.; Larner, J. M.; Helmke, B. P.; Sheng, K. Int. J. Radiat. Oncol. Biol. Phys.
Gazeau, F.; Wilhelm, C. ACS Nano 2013, 7, 4954. 2008, 72, 633.
(164) Sharma, S. K.; Chiang, L. Y.; Hamblin, M. R. Nanomedicine (195) Samia, A. C. S.; Chen, X.; Burda, C. J. Am. Chem. Soc. 2003,
(London) 2011, 6, 1813. 125, 15736.
(165) (a) Liao, F.; Saitoh, Y.; Miwa, N. Oncol. Res. 2011, 19, 203. (196) Shi, L.; Hernandez, B.; Selke, M. J. Am. Chem. Soc. 2006, 128,
(b) Tabata, Y.; Murakami, Y.; Ikada, Y. Jpn. J. Cancer Res. 1997, 88, 6278.
1108. (c) Liu, J.; Ohta, S.-i.; Sonoda, A.; Yamada, M.; Yamamoto, M.; (197) Tsay, J. M.; Trzoss, M.; Shi, L.; Kong, X.; Selke, M.; Jung, M.
Nitta, N.; Murata, K.; Tabata, Y. J. Controlled Release 2007, 117, 104. E.; Weiss, S. J. Am. Chem. Soc. 2007, 129, 6865.
(166) Ikeda, A.; Doi, Y.; Nishiguchi, K.; Kitamura, K.; Hashizume, (198) Hsu, C.-Y.; Chen, C.-W.; Yu, H.-P.; Lin, Y.-F.; Lai, P.-S.
M.; Kikuchi, J.-i.; Yogo, K.; Ogawa, T.; Takeya, T. Org. Biomol. Chem. Biomaterials 2013, 34, 1204.
2007, 5, 1158. (199) Saxena, V.; Sadoqi, M.; Shao, J. Int. J. Pharm. 2006, 308, 200.
(167) (a) Akiyama, M.; Ikeda, A.; Shintani, T.; Doi, Y.; Kikuchi, J.-i.; (200) Gomes, A. J.; Lunardi, L. O.; Marchetti, J. M.; Lunardi, C. N.;
Ogawa, T.; Yogo, K.; Takeya, T.; Yamamoto, N. Org. Biomol. Chem. Tedesco, A. C. Photomed. Laser Surg. 2006, 24, 514.

AY DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

(201) Gao, D.; Agayan, R. R.; Xu, H.; Philbert, M. A.; Kopelman, R. (229) Liu, X.; Zheng, M.; Kong, X.; Zhang, Y.; Zeng, Q.; Sun, Z.;
Nano Lett. 2006, 6, 2383. Buma, W. J.; Zhang, H. Chem. Commun. 2013, 49, 3224.
(202) Kim, S.; Ohulchanskyy, T. Y.; Pudavar, H. E.; Pandey, R. K.; (230) Zhang, P.; Steelant, W.; Kumar, M.; Scholfield, M. J. Am. Chem.
Prasad, P. N. J. Am. Chem. Soc. 2007, 129, 2669. Soc. 2007, 129, 4526.
(203) Velusamy, M.; Shen, J.-Y.; Lin, J. T.; Lin, Y.-C.; Hsieh, C.-C.; (231) Chatterjee, D. K.; Yong, Z. Nanomedicine (London) 2008, 3,
Lai, C.-H.; Lai, C.-W.; Ho, M.-L.; Chen, Y.-C.; Chou, P.-T.; Hsiao, J.- 73.
K. Adv. Funct. Mater. 2009, 19, 2388. (232) (a) Ungun, B.; Prud’homme, R. K.; Budijon, S. J.; Shan, J.;
(204) Gary-Bobo, M.; Mir, Y.; Rouxel, C.; Brevet, D.; Basile, I.; Lim, S. F.; Ju, Y.; Austin, R. Opt. Express 2009, 17, 80. (b) Wang, C.;
Maynadier, M.; Vaillant, O.; Mongin, O.; Blanchard-Desce, M.; Tao, H.; Cheng, L.; Liu, Z. Biomaterials 2011, 32, 6145.
Morère, A.; Garcia, M.; Durand, J.-O.; Raehm, L. Angew. Chem., Int. (233) Shan, J.; Chen, J.; Meng, J.; Collins, J.; Soboyejo, W.;
Ed. 2011, 50, 11425. Friedberg, J. S.; Ju, Y. J. Appl. Phys. 2008, 104, 094308.
(205) Dayal, S.; Burda, C. J. Am. Chem. Soc. 2008, 130, 2890. (234) (a) Cui, S.; Chen, H.; Zhu, H.; Tian, J.; Chi, X.; Qian, Z.;
(206) Wen, Y.-n.; Song, W.-s.; An, L.-m.; Liu, Y.-q.; Wang, Y.-h.; Achilefu, S.; Gu, Y. J. Mater. Chem. 2012, 22, 4861. (b) Zhou, A.; Wei,
Yang, Y.-q. Appl. Phys. Lett. 2009, 95. Y.; Wu, B.; Chen, Q.; Xing, D. Mol. Pharmaceutics 2012, 9, 1580.
(207) Qi, Z.-D.; Li, D.-W.; Jiang, P.; Jiang, F.-L.; Li, Y.-S.; Liu, Y.; (c) Cui, S.; Yin, D.; Chen, Y.; Di, Y.; Chen, H.; Ma, Y.; Achilefu, S.;
Wong, W.-K.; Cheah, K.-W. J. Mater. Chem. 2011, 21, 2455. Gu, Y. ACS Nano 2013, 7, 676.
(208) Skripka, A.; Valanciunaite, J.; Dauderis, G.; Poderys, V.; (235) (a) Guo, H.; Qian, H.; Idris, N. M.; Zhang, Y. Nanomedicine
Kubiliute, R.; Rotomskis, R. J. Biomed. Opt. 2013, 18, 078002. 2010, 6, 486. (b) Idris, N. M.; Gnanasammandhan, M. K.; Zhang, J.;
(209) Shen, X.; He, F.; Wu, J.; Xu, G. Q.; Yao, S. Q.; Xu, Q.-H. Ho, P. C.; Mahendran, R.; Zhang, Y. Nat. Med. 2012, 18, 1580.
Langmuir 2011, 27, 1739. (c) Qian, H. S.; Guo, H. C.; Ho, P. C.; Mahendran, R.; Zhang, Y. Small
(210) Shen, X.; Li, L.; Wu, H.; Yao, S. Q.; Xu, Q.-H. Nanoscale 2011, 2009, 5, 2285.
3, 5140. (236) Shan, J.; Budijono, S. J.; Hu, G.; Yao, N.; Kang, Y.; Ju, Y.;
(211) Zhao, T.; Yu, K.; Li, L.; Zhang, T.; Guan, Z.; Gao, N.; Yuan, P.; Prud’homme, R. K. Adv. Funct. Mater. 2011, 21, 2488.
Li, S.; Yao, S. Q.; Xu, Q.-H.; Xu, G. Q. ACS Appl. Mater. Interfaces (237) Wang, H.; Liu, Z.; Wang, S.; Dong, C.; Gong, X.; Zhao, P.;
2014. Chang, J. ACS Appl. Mater. Interfaces 2014, 6, 3219.
(212) Ogawa, K.; Kobuke, Y. Anticancer Agents Med. Chem. 2008, 8, (238) (a) Xu, Q. C.; Zhang, Y.; Tan, M. J.; Liu, Y.; Yuan, S.; Choong,
269. C.; Tan, N. S.; Tan, T. T. Adv. Healthc. Mater. 2012, 1, 470. (b) Liu,
(213) Boyer, J. C.; Vetrone, F.; Cuccia, L. A.; Capobianco, J. A. J. Am. K.; Liu, X.; Zeng, Q.; Zhang, Y.; Tu, L.; Liu, T.; Kong, X.; Wang, Y.;
Chem. Soc. 2006, 128, 7444. Cao, F.; Lambrechts, S. A.; Aalders, M. C.; Zhang, H. ACS Nano 2012,
(214) Auzel, F. Chem. Rev. 2004, 104, 139. 6, 4054.
(215) Yen, W. M., Weber, M. J. ; Inorganic Phosphors: Compositions, (239) (a) Aveline, B. M.; Hasan, T.; Redmond, R. W. J. Photochem.
Preparation, and Optical Properties; CRC Press: Boca Raton, FL, 2004. Photobiol., B 1995, 30, 161. (b) Kuznetsova, N. A.; Gretsova, N. S.;
(216) Wang, F.; Liu, X. Chem. Soc. Rev. 2009, 38, 976. Derkacheva, V. M.; Kaliya, O. L.; Lukyanets, E. A. J. Porphyrins
(217) (a) Rriedener, T.; Kraemer, K.; Guedel, H. U. ChemInform Phthalocyanines 2003, 07, 147. (c) Tanielian, C.; Schweitzer, C.;
1995, 26. (b) Suyver, J. F.; Aebischer, A.; Biner, D.; Gerner, P.; Mechin, R.; Wolff, C. Free Radical Biol. Med. 2001, 30, 208.
Grimm, J.; Heer, S.; Krämer, K. W.; Reinhard, C.; Güdel, H. U. Opt. (240) Wang, M.; Chen, Z.; Zheng, W.; Zhu, H.; Lu, S.; Ma, E.; Tu,
Mater. 2005, 27, 1111. D.; Zhou, S.; Huang, M.; Chen, X. Nanoscale 2014, 6, 8274.
(218) (a) Patra, A.; Friend, C. S.; Kapoor, R.; Prasad, P. N. Appl. (241) Idris, N. M.; Lucky, S. S.; Li, Z.; Huang, K.; Zhang, Y. J. Mater.
Phys. Lett. 2003, 83, 284. (b) Yi, G.; Lu, H.; Zhao, S.; Ge, Y.; Yang, W.; Chem. B 2014, 2, 7017.
Chen, D.; Guo, L.-H. Nano Lett. 2004, 4, 2191. (242) Lucky, S. S.; Idris, N. M.; Li, Z.; Huang, K.; Soo, K. C.; Zhang,
(219) (a) Krämer, K. W.; Biner, D.; Frei, G.; Güdel, H. U.; Hehlen, Y. ACS Nano, in press.
M. P.; Lüthi, S. R. Chem. Mater. 2004, 16, 1244. (b) Schäfer, H.; (243) Wang, F.; Chatterjee, D. K.; Li, Z.; Zhang, Y.; Fan, X.; Wang,
Ptacek, P.; Eickmeier, H.; Haase, M. Adv. Funct. Mater. 2009, 19, 3091. M. Nanotechnology 2006, 17, 5786.
(c) Yi, G.-S.; Chow, G.-M. Chem. Mater. 2006, 19, 341. (244) Chen, Z.; Chen, H.; Hu, H.; Yu, M.; Li, F.; Zhang, Q.; Zhou,
(220) Jin, S.; Zhou, L.; Gu, Z.; Tian, G.; Yan, L.; Ren, W.; Yin, W.; Z.; Yi, T.; Huang, C. J. Am. Chem. Soc. 2008, 130, 3023.
Liu, X.; Zhang, X.; Hu, Z.; Zhao, Y. Nanoscale 2013, 5, 11910. (245) Li, Z.; Zhang, Y. Nanotechnology 2008, 19, 345606.
(221) Suyver, J. F.; Grimm, J.; van Veen, M. K.; Biner, D.; Krämer, K. (246) Liu, C.; Wang, H.; Li, X.; Chen, D. J. Mater. Chem. 2009, 19,
W.; Güdel, H. U. J. Lumin. 2006, 117, 1. 3546.
(222) (a) Zhao, Z.; Han, Y.; Lin, C.; Hu, D.; Wang, F.; Chen, X.; (247) Park, Y. I.; Kim, J. H.; Lee, K. T.; Jeon, K.-S.; Na, H. B.; Yu, J.
Chen, Z.; Zheng, N. Chem.Asian J. 2012, 7, 830. (b) Qiao, X. F.; H.; Kim, H. M.; Lee, N.; Choi, S. H.; Baik, S.-I.; Kim, H.; Park, S. P.;
Zhou, J. C.; Xiao, J. W.; Wang, Y. F.; Sun, L. D.; Yan, C. H. Nanoscale Park, B.-J.; Kim, Y. W.; Lee, S. H.; Yoon, S.-Y.; Song, I. C.; Moon, W.
2012, 4, 4611. (c) Wang, X.; Liu, K.; Yang, G.; Cheng, L.; He, L.; Liu, K.; Suh, Y. D.; Hyeon, T. Adv. Mater. 2009, 21, 4467.
Y.; Li, Y.; Guo, L.; Liu, Z. Nanoscale 2014. (248) Wang, Y.; Tu, L.; Zhao, J.; Sun, Y.; Kong, X.; Zhang, H. J. Phys.
(223) Park, Y. I.; Kim, H. M.; Kim, J. H.; Moon, K. C.; Yoo, B.; Lee, Chem. C 2009, 113, 7164.
K. T.; Lee, N.; Choi, Y.; Park, W.; Ling, D.; Na, K.; Moon, W. K.; (249) Wang, F.; Fan, X.; Wang, M.; Zhang, Y. Nanotechnology 2007,
Choi, S. H.; Park, H. S.; Yoon, S. Y.; Suh, Y. D.; Lee, S. H.; Hyeon, T. 18, 025701.
Adv. Mater. 2012. (250) Wang, F.; Liu, X. J. Am. Chem. Soc. 2008, 130, 5642.
(224) Bunzli, J.-C. G.; Piguet, C. Chem. Soc. Rev. 2005, 34, 1048. (251) Tian, G.; Gu, Z.; Zhou, L.; Yin, W.; Liu, X.; Yan, L.; Jin, S.;
(225) BG, W. Spectroscopic properties of rare earths; Interscience Ren, W.; Xing, G.; Li, S.; Zhao, Y. Adv. Mater. 2012, 24, 1226.
Publishers: New York, 1965. (252) Zeng, L.; Xiang, L.; Ren, W.; Zheng, J.; Li, T.; Chen, B.; Zhang,
(226) (a) Jiang, C.; Xu, W. J. Display Technol. 2009, 5, 312. (b) Chen, J.; Mao, C.; Li, A.; Wu, A. R. Soc. Chem. Adv. 2013, 3, 13915.
F.; Zhang, S.; Bu, W.; Chen, Y.; Xiao, Q.; Liu, J.; Xing, H.; Zhou, L.; (253) Wang, H. J.; Shrestha, R.; Zhang, Y. Part. Part. Syst. Charact.
Peng, W.; Shi. Chem.Eur. J. 2012, 18, 7082. 2014, 31, 228.
(227) (a) Tian, G.; Ren, W.; Yan, L.; Jian, S.; Gu, Z.; Zhou, L.; Jin, S.; (254) Liu, Q.; Sun, Y.; Yang, T.; Feng, W.; Li, C.; Li, F. J. Am. Chem.
Yin, W.; Li, S.; Zhao, Y. Small 2013, 9, 1929. (b) Wang, C.; Cheng, L.; Soc. 2011, 133, 17122.
Liu, Y.; Wang, X.; Ma, X.; Deng, Z.; Li, Y.; Liu, Z. Adv. Funct. Mater. (255) Yuan, Q.; Wu, Y.; Wang, J.; Lu, D.; Zhao, Z.; Liu, T.; Zhang,
2013, 23, 3077. X.; Tan, W. Angew. Chem., Int. Ed. 2013, 52, 13965.
(228) Xia, L.; Kong, X.; Liu, X.; Tu, L.; Zhang, Y.; Chang, Y.; Liu, K.; (256) Chen, Q.; Wang, C.; Cheng, L.; He, W.; Cheng, Z.; Liu, Z.
Shen, D.; Zhao, H.; Zhang, H. Biomaterials 2014, 35, 4146. Biomaterials 2014, 35, 2915.

AZ DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

(257) Wang, X.; Yang, C.-X.; Chen, J.-T.; Yan, X.-P. Anal. Chem. Fafarman, A. T.; Chettiar, U. K.; Engheta, N.; Murray, C. B.; Kagan, C.
2014, 86, 3263. R. ACS Nano 2012, 6, 8758.
(258) Wang, H.; Dong, C.; Zhao, P.; Wang, S.; Liu, Z.; Chang, J. Int. (286) Zhan, Q.; Qian, J.; Liang, H.; Somesfalean, G.; Wang, D.; He,
J. Pharm. 2014, 466, 307. S.; Zhang, Z.; Andersson-Engels, S. ACS Nano 2011, 5, 3744.
(259) Li, Z. Q. a. Z. Y. Angew. Chem., Int. Edn. 2006, 45, 7732. (287) Zou, W.; Visser, C.; Maduro, J. A.; Pshenichnikov, M. S.;
(260) Wang, H.; Wang, S.; Liu, Z.; Dong, C.; Yang, J.; Gong, X.; Hummelen, J. C. Nat. Photonics 2012, 6, 560.
Chang, J. Nanotechnology 2014, 25, 155103. (288) (a) Wen, H.; Zhu, H.; Chen, X.; Hung, T. F.; Wang, B.; Zhu,
(261) Wang, F.; Han, Y.; Lim, C. S.; Lu, Y.; Wang, J.; Xu, J.; Chen, G.; Yu, S. F.; Wang, F. Angew. Chem., Int. Ed. 2013, 52, 13419. (b) Li,
H.; Zhang, C.; Hong, M.; Liu, X. Nature 2010, 463, 1061. X.; Wang, R.; Zhang, F.; Zhou, L.; Shen, D.; Yao, C.; Zhao, D. Sci. Rep.
(262) Yuan, Y.; Min, Y.; Hu, Q.; Xing, B.; Liu, B. Nanoscale 2014, 6, 2013, 3.
11259. (289) Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U. S.
(263) Chen, F.; Bu, W.; Zhang, S.; Liu, X.; Liu, J.; Xing, H.; Xiao, Q.; Angew. Chem., Int. Ed. 2010, 49, 6288.
Zhou, L.; Peng, W.; Wang, L.; Shi, J. Adv. Funct. Mater. 2011, 21, 4285. (290) (a) Wang, M.; Thanou, M. Pharmacol. Res. 2010, 62, 90.
(264) Fan, W.; Shen, B.; Bu, W.; Chen, F.; He, Q.; Zhao, K.; Zhang, (b) Ferrari, M. Nat. Nanotechnol. 2008, 3, 131.
S.; Zhou, L.; Peng, W.; Xiao, Q.; Ni, D.; Liu, J.; Shi, J. Biomaterials
2014, 35, 8992.
(265) Chen, G.; Shen, J.; Ohulchanskyy, T. Y.; Patel, N. J.; Kutikov,
A.; Li, Z.; Song, J.; Pandey, R. K.; Ågren, H.; Prasad, P. N.; Han, G.
ACS Nano 2012, 6, 8280.
(266) Punjabi, A.; Wu, X.; Tokatli-Apollon, A.; El-Rifai, M.; Lee, H.;
Zhang, Y.; Wang, C.; Liu, Z.; Chan, E. M.; Duan, C.; Han, G. ACS
Nano 2014.
(267) Nesmerak, K. In Encyclopedia of Metalloproteins; Kretsinger, R.,
Uversky, V., Permyakov, E., Eds.; Springer: New York, 2013.
(268) (a) Chen, J.; Guo, C.; Wang, M.; Huang, L.; Wang, L.; Mi, C.;
Li, J.; Fang, X.; Mao, C.; Xu, S. J. Mater. Chem. 2011, 21, 2632.
(b) Zhou, J.-C.; Yang, Z.-L.; Dong, W.; Tang, R.-J.; Sun, L.-D.; Yan,
C.-H. Biomaterials 2011, 32, 9059.
(269) Abdul Jalil, R.; Zhang, Y. Biomaterials 2008, 29, 4122.
(270) Xiong, L.; Yang, T.; Yang, Y.; Xu, C.; Li, F. Biomaterials 2010,
31, 7078.
(271) Cheng, L.; Yang, K.; Shao, M.; Lu, X.; Liu, Z. Nanomedicine
2011, 6, 1327.
(272) Xiong, L.; Chen, Z.; Tian, Q.; Cao, T.; Xu, C.; Li, F. Anal.
Chem. 2009, 81, 8687.
(273) Xiong, L.-Q.; Chen, Z.-G.; Yu, M.-X.; Li, F.-Y.; Liu, C.; Huang,
C.-H. Biomaterials 2009, 30, 5592.
(274) Wang, S.; Gao, R.; Zhou, F.; Selke, M. J. Mater. Chem. 2004,
14, 487.
(275) Yang, L.; Alexandridis, P. Curr. Opin. Colloid Interface Sci. 2000,
5, 132.
(276) (a) Emerich, D. F.; Thanos, C. G. Biomol. Eng. 2006, 23, 171.
(b) Zamboni, W. C. Oncologist 2008, 13, 248.
(277) (a) Powers, K. W.; Brown, S. C.; Krishna, V. B.; Wasdo, S. C.;
Moudgil, B. M.; Roberts, S. M. Toxicol. Sci. 2006, 90, 296.
(b) Murdock, R. C.; Braydich-Stolle, L.; Schrand, A. M.; Schlager, J.
J.; Hussain, S. M. Toxicol. Sci. 2008, 101, 239.
(278) (a) Braathen, L. R.; Szeimies, R. M.; Basset-Seguin, N.;
Bissonnette, R.; Foley, P.; Pariser, D.; Roelandts, R.; Wennberg, A. M.;
Morton, C. A. J. Am. Acad. Dermatol. 2007, 56, 125. (b) Niemz, M. H.
Laser-Tissue Interactions:Fundamentals and Applications, 3rd ed.;
Springer: Hiedelberg, Germany, 2007.
(279) Boyer, J. C.; van Veggel, F. C. Nanoscale 2010, 2, 1417.
(280) Cheng, Q.; Sui, J.; Cai, W. Nanoscale 2012, 4, 779.
(281) Ramasamy, P.; Chandra, P.; Rhee, S. W.; Kim, J. Nanoscale
2013, 5, 8711.
(282) (a) Shen, J.; Chen, G.; Vu, A.-M.; Fan, W.; Bilsel, O. S.; Chang,
C.-C.; Han, G. Adv. Opt. Mater. 2013, 1, 644. (b) Heer, S.; Kömpe, K.;
Güdel, H. U.; Haase, M. Adv. Mater. 2004, 16, 2102. (c) Wang, L.;
Lan, M.; Liu, Z.; Qin, G.; Wu, C.; Wang, X.; Qin, W.; Huang, W.;
Huang, L. J. Mater. Chem. C 2013, 1, 2485.
(283) (a) Pan, L.; He, M.; Ma, J.; Tang, W.; Gao, G.; He, R.; Su, H.;
Cui, D. Theranostics 2013, 3, 210. (b) Zeng, S.; Xiao, J.; Yang, Q.; Hao,
J. J. Mater. Chem. 2012, 22, 9870.
(284) (a) Zeng, Q.; Xue, B.; Zhang, Y.; Wang, D.; Liu, X.; Tu, L.;
Zhao, H.; Kong, X.; Zhang, H. CrystEngComm 2013, 15, 4765.
(b) Chen, M.; Ma, Y.; Li, M. Mater. Lett. 2014, 114, 80.
(285) (a) Jiang, T.; Liu, Y.; Liu, S.; Liu, N.; Qin, W. J. Colloid Interface
Sci. 2012, 377, 81. (b) Saboktakin, M.; Ye, X.; Oh, S. J.; Hong, S. H.;

BA DOI: 10.1021/cr5004198
Chem. Rev. XXXX, XXX, XXX−XXX

View publication stats

S-ar putea să vă placă și