3. Conquering Multidrug Resistant Lung Cancer by Upconversion Nanoparticles‐Mediated

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Received: 1 January 2022 Accepted: 7 March 2022

DOI: 10.1002/jccs.202200001

ARTICLE

Conquering multidrug resistant lung cancer by


upconversion nanoparticles-mediated photodynamic
therapy and gene silencing

Poliraju Kalluru1 | Munusamy Shanmugam1 | Raviraj Vankayala2 |


Chi-Shiun Chiang3 | Kuo Chu Hwang1

1
Department of Chemistry, National Tsing
Hua University, Hsinchu, Taiwan, R.O.C.
Abstract
2
Department of Bioscience and Multidrug resistance (MDR) problem is a challenging task in cancer treat-
Bioengineering, Indian Institute of ments. Among various types of cancers, lung cancer is considered incurable, as
Technology Jodhpur, Jodhpur, Rajasthan,
it is diagnosed very often at the advanced metastatic stage, where surgical
India
3
Department of Biomedical Engineering
operations and radiation treatments become highly ineffective. In addition,
and Environmental Sciences, National lung cancers are multidrug resistant either intrinsically or upon continuous
Tsing Hua University, Hsinchu, Taiwan, administration and stimulation of chemotherapeutic drugs. To conquer the dif-
R.O.C.
ficult MDR problem, in this study, we developed a simple and effective thera-
Correspondence peutic strategy to achieve complete destruction of multidrug resistant lung
Kuo Chu Hwang, Department of
cancer by an unprecedented upconversion nanoparticles (UCNPs)-mediated
Chemistry, National Tsing Hua
University, Hsinchu 30013, Taiwan, photodynamic therapy (PDT) combined with effective (~70%) superoxide dis-
R.O.C. mutase 1 (SOD1) gene silencing using ultra-low doses (320 mW/cm2) of NIR
Email: kchwang@mx.nthu.edu.tw
light (980 nm) excitation. To the best of our knowledge, this is the first success-
Funding information ful literature example in achieving complete destruction of multidrug resistant
Frontier Research Center on Fundamental cancers. Overall, the current work points out a new direction to the clinicians
and Applied Sciences of Matters, National
Tsing Hua University, Taiwan; Ministry of
for the therapeutic design and effective treatments of multidrug resistant
Science and Technology, Taiwan cancers.

KEYWORDS
gene silencing, multidrug resistance, near infrared light, photodynamic therapy,
upconversion nanoparticles

1 | INTRODUCTION before it reaches the target intracellular organelle.[2,3]


MDR problem is commonly observed in various types of
Multidrug resistance (MDR) is one of the major obstacles cancers. Among them, lung cancer is considered incur-
by which many cancers develop resistance toward molec- able, because the survival rate of lung cancer patients is
ular anticancer drugs, leading to the failure of chemo- very low (<15%). The majority of the lung cancer patients
therapeutic treatments and deaths.[1–4] MDR generally were very often diagnosed only at the advanced meta-
arises due to stimulation of chemotherapeutic drugs and static stage, where the surgical operation and radiation
subsequent overexpression of cellular membrane pro- treatments are usually ineffective.[4,5] The non-small cell
teins, such as, ATP-binding cassette (ABC) transporter, p- lung cancers (NSCLC) was found to be intrinsically
glycoprotein (p-gp), and multidrug resistance associated multidrug resistant with overexpression of MRP family
proteins (MRPs), that can mediate molecular drug efflux proteins; but not the p-gp.[6] Previously, a lot of efforts

J Chin Chem Soc. 2022;1–13. http://www.jccs.wiley-vch.de © 2022 The Chemical Society Located in Taipei & Wiley-VCH GmbH 1
2 KALLURU ET AL.

have been devoted to overcoming the MDR problem of effective cancer cell deaths.[41–43] In addition to the SOD1
lung tumors, but only partial suppression or delay tumor gene, there are still many other genes available, such as
growths were achieved.[7–10] Therefore, it leaves a grand survivin, polo-like kinase (Plk1), Bcl-2, tumor necrosis
challenge to develop a noninvasive therapeutic modality factor (TNF-α), HSP70, and HSPA1A, which can help the
that can effectively conquer the MDR problem, and facili- cancer cells to prevent themselves from undergoing oxi-
tate complete destruction of lung tumors. dation stress-induced deaths.[44–47] These genes are
In addition to the chemotherapy, radiotherapy and pho- potential targets for gene silencing to suppress cancer
totherapy are two treatment modalities that can cure cancer cells' ability against oxidation stress.
effectively. Among them, phototherapeutic approach has In the current work, we present an unprecedented,
many advantages such as, noninvasiveness,[11] excellent simple, but effective therapeutic strategy to achieve the
spatial–temporal selectivity, low side effects,[12] and complete destruction of multidrug resistant cancers/
cost-effective as compared to other treatment modalities.[13] tumors. We develop nanoparticles prodrug to generate
Photodynamic therapy (PDT) requires the use of organic reactive oxygen species (ROS) to kill cancer cells having
photosensitizers (PS) to absorb and transfer the photon MDR proteins. We foresee that multidrug resistant pro-
energy to molecular oxygen (3O2) in tumor tissues, to gener- teins cannot pump out large (relative to molecular drugs)
ate cytotoxic singlet oxygen (1O2) and kill cancer cells.[14] nanoparticles-based nanomedicines,[40] which are very
Conventional organic PS-mediated PDT has many limita- different from small chemodrugs in terms of sizes, sur-
tions, such as, poor water solubility, severe photo-bleaching face functionalities and polarities. Therefore, cancer cells
problem, restriction to visible light activation, which has with MDR proteins can be easily killed the same as those
very short tissue-penetration depths, and therefore not without multidrug resistant abilities. Our in vivo lung
applicable to deep tissue-buried tumors. To treat deep cancer experiments demonstrate that such a therapeutic
tissue-buried tumors, near infrared (NIR) light with far design works very well and solid lung tumor with intrin-
larger tissue penetration depths must be used. However, sic MDR proteins can be completely destroyed.
NIR light activatable organic PS molecules are very rare.[15]
Currently, the use of nanomaterials to absorb NIR light to
exert NIR PDT is becoming the major trend of photother- 2 | EXPERIMENTAL SECTION
apy for cancer treatment.[16] Previously, upconversion
nanoparticles (UCNPs) were used to absorb and upconvert 2.1 | Materials or reagents
NIR light to visible/uv light for indirect sensitization of
organic photosensitizers for treatments of non-multidrug Yttrium oxide (99.9%), ytterbium oxide (99.9%), and erbium
resistant tumors.[17–21] However, only partial suppression or oxide (99.9%), hydrochloric acid, oleic acid, octadecene,
delay the growth of tumors was achieved, albeit very high sodium hydroxide, 1-ethyl-3-(3-dimethylaminopropyl)
laser powers (0.33 ~ 0.78 W/cm2) of NIR (980 nm) light was carbodiimide (EDC), and N-Hydroxysulfosuccinimide (Sul-
used to photochemically excite the UCNPs, which is proba- NHS) were purchased from Sigma-Aldrich, USA. Ammo-
bly due to multiple-step sensitization and therefore nium fluoride was purchased from Alfa-Aesar, USA,
extremely low overall efficiencies of singlet O2 whereas methanol is from Echo Chemicals Co. Ltd.,
generation.[22–26] Multiple-step photosensitization processes Taiwan. Anti-epidermal growth factor receptor antibody
always suffer from very low overall quantum efficiency for (anti-EGFR) was obtained from R&D systems, USA.
singlet oxygen generation. Recently, we have reported sev- 40 ,6-Diamidino-2-phenylindole-dihydrochloride (DAPI) was
eral examples of near infrared light initiated photodynamic obtained from Thermo Fisher Scientific, USA.
therapy (NIR-PDT) for effective treatments for non-
multidrug resistant cancers, such as, gold nanorods,
nanoshells, nanoechinus, and tungsten oxide nanowires, 2.2 | Synthesis of UCNPs
can sensitize formation of singlet oxygen directly by NIR
light irradiation, and simultaneously exert PDT effects for In a typical experiment, Y-oleate (0.68 mmol), Yb-oleate
the destruction of tumors without drug resistant capability (0.3 mmol), and Er-oleate (0.02 mmol) complexes were
under low laser doses (<0.4 W/cm2).[27–40] mixed with 6 ml of oleic acid and 15 ml 1-octadecene in a
In the literature, it is known that the SOD1 gene is an 100 ml three neck round bottomed flask. The reaction
essential enzyme that eliminates superoxide radical mixture was refluxed at 150 C in an inert Ar atmosphere
(O2.) and thus protects cancer cells from ROS-induced until getting clear solution and then cooled down to room
damages. Silencing of the SOD1 gene of cancer cells is temperature. Then, a 10 ml of a methanol solution con-
expected to suppress the antioxidation ability of cancer taining NaOH (2.5 mmol) and NH4F (4 mmol) was gently
cells and thus trigger apoptosis efficiently, leading to the added to the reaction mixture and stirred for 20 min at
KALLURU ET AL. 3

RT. Then, methanol was removed by vacuum distillation. a luminescence spectrometer (FLS920, Edinburgh Instru-
The reaction mixture was heated at 315 C for 90 min. ments Ltd. Scotland, UK). A 1000 nm long pass filter
The resulting reaction mixture was then immediately (Iszu Optics, Taiwan) was placed in-between the light
cooled down to room temperature. Upon addition of eth- source and detector to avoid the stray and scattering light
anol into the reaction mixture, the UCNPs were precipi- from the light source which is a 450 W Xe lamp.
tated and then collected upon ultra-centrifugation. The
contents were centrifuged for 3 ~ 5 times to remove
excess surfactant and solvent. The purified UCNPs were 2.6 | DPBF quenching experiment
then dispersed in hexane.
0.08 mM DPBF (1,3-diphenyl isobenzofuran) and
1 mg/ml of citrate-UCNPs were thoroughly mixed and
2.3 | Synthesis of citrate-capped UCNPs irradiated with 980 nm laser with the power intensity of
320 mW/cm2 for 30 min. The absorbance of the solution
Thirty milligrams of hydrophobic UCNPs (NaYF4: Yb, at 410 nm was measured for every 5 min using a
Er) was mixed with 3 ml of chloroform and 2 ml of tolu- UV–visible spectrophotometer. The decrease in the
ene, followed by ultrasonication. This suspension solu- absorbance of DPBF caused by the photobleaching was
tion was slowly injected into the three-necked flask measured in all experiments.
containing 150 mg of citric acid and 15 ml of diethylene
glycol, and heated at 110 C within 1.5 hr under argon
atmosphere. Then, air was introduced into the solution, 2.7 | EPR measurements
which was further heated at 160 C for another 1.5 hr
until the solution becomes clear. The reaction was cooled For the detection of 1O2 using EPR, 500 μl of 1 mg/ml
down to room temperature and the solid contents were citrate-UCNPs in D2O suspension was added to the 40 μl
washed thoroughly with ethanol and deionized of 0.3 M TEMP in D2O solution, and the resulting solu-
(DI) water. The citrate-capped UCNPs were finally re- tion was mixed well. The mixture was then bubbled with
suspended in DI water to obtain a solution with a final O2 for 5 min and then irradiated with 980 nm laser with
UCNP concentration of 1.0 mg/ml. the power intensity of 320 mW/cm2 right in front of the
ESR spectrometer during the measurements. 10 mM his-
tidine was used as a scavenger for 1O2 in these experi-
2.4 | Synthesis of anti-EGFR conjugated ments. Parameter settings: microwave power, 10.12 mW;
UCNPs frequency, 9.8 GHz; time constant, 40.96 ms; scan
width, 100 G.
One milligram of citrate-capped UCNPs were mixed in
500 μl phosphate-buffered saline (PBS, pH ~ 7.4), followed
by the addition of a solution containing 1.5 mg of 1-ethyl- 2.8 | Cell culture, materials, and
3-(3-dimethylaminopropyl) carbodiimide (EDC) and 1 mg reagents
of N-hydroxysulfosuccinimide (Sul-NHS), and vortex for
1 min. Further, 20 μl (10 μg) of anti- EGFR was added and Non-small cell lung cancer (NCI-H23) cells were grown in
stirred for overnight at 4 C. Later, 1 ml PBS was added Roswell Park Memorial Institute medium (RPMI-1640)
and then centrifuged at 10000 rpm for 5–6 min. The anti- (Gibco, USA) supplemented with 10% heat-inactivated
EGFR conjugated UCNPs settle down to the bottom of the fetal bovine serum (Gibco, USA), 2 mM L-glutamine,
eppendorf tube, where the supernatant contains the 100 μg/ml penicillin, and 100 U/ml streptomycin.
unreacted anti-EGFR. Upon washing for 3 times, the The cells were grown in a humidified incubator at 37 C
anti-EGFR conjugated UCNPs were used for further (95% humidity, 5% CO2).
experiments.

2.9 | In vitro uptake studies


2.5 | Singlet oxygen phosphorescence
emission measurements For confocal microscopy experiments, NCI-H23 cells
(2  105 cells/ml) were cultured in 6-well dish with
Citrate capped-UCNPs (1 mg/ml) were dispersed in D2O RPMI-1640 medium, and then added different concentra-
solvent, followed by photo excitation to generate singlet tions of anti-EGFR-UCNPs and incubated for 24 hr. After
O2 phosphorescence emission, which was recorded using that 1x phosphate buffered saline (PBS) solution was
4 KALLURU ET AL.

used to wash the cells. The cells were then further treated solution was further centrifuged for 10 min
by 4% paraformaldehyde solution on to the glass slide (at 12000 rpm) at 4 C. 200 μl of chloroform was then
and then followed by PBS wash and once again treated added into it, vortexed for 20 s, and then kept it stand by
with 5% Tween-20 in PBS solution onto a glass slide and in ice for 10 min. The solution was then centrifuged at
then further two times washed with PBS. The clean 12000 rpm for 20 min at 4 C and RNA was obtained in
washed NCI-H23 cells were stained with DAPI the supernatant which was collected into another clean
(40 ,6-diamidino-2-phenylindole) and observed under a eppendorf tube. Now equal volume of 2-propanol was
multi-photon confocal scanning microscope (Leica TCS added to precipitate the RNA and kept at 20 C for
SPX5, Germany), which was equipped with a 404 nm uv 24 hr. Later, the contents were centrifuged once again at
laser and a tunable NIR laser. For all the uptake experi- 12000 rpm for 20 min at 4 C and the supernatant was
ments, 980 nm excitation light was used with a laser removed. To the bottom pellet, 1 ml of 75% ethanol was
power intensity of 5 mW/cm2. The emission was col- added, vortexed, and centrifuged for 20 min at 4 C.
lected from the PI (propidium iodide) channel Finally, an appropriate volume of RNAse free water was
(λem = 600–700 nm). added to the air-dried RNA pellet and stored at 20 C
for further experiments. The final concentration of total
RNA was measured using Nanodrop.
2.10 | Preparation of siRNA-anti-EGFR-
UCNPs
2.13 | Polymerase chain reaction (PCR)
A stock solution of 0.5 μM of superoxide dismutase of total RNA to complementary DNA
1 (SOD1) siRNA (siGENOME Human SOD1 [6647],
siRNA SMART pool, Thermo Scientific, USA) was Two micrograms of total RNA was taken in a neat
diluted to 100 nM using RNAse free water. Then the eppendorf tube and various PCR components were added
stock solution of anti-EGFR-UCNPs (1 mg/ml) was in a sequence as follows. First 5 mM dNTP mix
added to the dilute siRNA solution and vortexed for few (Promega, USA) was added, followed by the addition of
minutes and kept undisturbed for 15 min at room tem- 10 μM oligo-dT primer (Thermo Fisher Scientific, USA)
perature to form a stable complex before use. and vortexed gently. In the next step, 10 units/μl of
RNAse inhibitor (Promega, USA) was added, which was
followed by the addition of Omniscript Reverse Tran-
2.11 | Size and zeta-potential scriptase (Promega, USA). The eppendorf tube was kept
measurements at 37 C for 1 hr to complete the PCR reaction. The as
obtained complementary DNA (cDNA) concentration
The siRNA-anti-EGFR-UCNPs complexes were prepared was measured using Nanodrop Lite spectrophotometer
accordingly from the abovementioned procedures. The (ThermoFisher Scientific, USA) and used for further
surface charge and average particle size was measured experiments.
using Zeta-sizer instrument (Malvern, UK).

2.14 | Real-time quantitative PCR (RT-


2.12 | Isolation of total RNA from qPCR) experiment
siRNA-anti-EGFR-UCNPs internalized
NCI-H23 cells The cDNA was served as a template and the manufacturer
protocol was followed for the RT-qPCR experiment (SYBR
NCI-H23 cells were cultured in a 6-well plate with a Green Master Mix, Thermo Scientific, USA). In a typical
seeding density of 2.0  105 cells per well and incubated experiment, an appropriate concentration of cDNA
for 24 hr. The cells were treated with different concentra- (~500 ng) was added into the PCR tube. In the next step,
tions of siRNA-anti-EGFR-UCNPs nanoconjugates for the forward and reverse primers (~0.3 μM each) for the
24 hr. The cells were then washed with a phosphate target gene (SOD1, 50 -TGTGGCCGATGTGTCTATTG-30
buffer solution (PBS, pH 7.4) and incubated for an addi- (forward) and 50 -GCGTTTCCTGTCTTTGTACTTTC-30
tional time of 24 hr. Further, the cells were washed with (reverse)) and house-keeping gene (GADPH, 50 -TGAA
PBS for one more time, trypsinized and followed by cen- GGTCGG-AGTCAACGGATTTGGT-30 (forward) and 50 -
trifugation to make a cell pellet. Then 1 ml of TRIZOL CATGTGGGCCATGAGGTCCACCAC-30 (reverse)) were
(Sigma-Aldrich, USA) reagent was added to the cell pellet also added and gently pipetted to ensure thorough mixing
and the contents were kept at 80 C for 24 hr. The of the components. During the last step, 12.5 μl of SYBR
KALLURU ET AL. 5

Green dye was added, followed by the addition of RNAse 2.17 | Singlet oxygen sensor green
free water to make a total volume of 25 μl. The tubes were (SOSG) experiments
kept in the RT-qPCR machine (Thermo Scientific, USA)
with the following steps: (a) Uracil-DNA glycolase (UDG) NCI-H23 cells were loaded into the 6-well dishes with
pre-treatment at 50 C for 2 min; (b) 1 cycle of initial dena- the density of 2  105 cells per well. After that incubate
turation at 95 C for 10 min; (c) 1 cycle of denaturation at for 24 hr, anti-EGFR-UCNPs, and siRNA-anti-EGFR-
95 C for 15 s; and (d) 40 cycles of annealing/extension at UCNPs stock solutions were added to the cells, followed
60 C for 60 s. The results were analyzed using the ΔΔCT by incubation for 24 hr. Then, the medium was added
method (comparative CT method) where CT is the thresh- 2 μl of singlet oxygen sensor probe SOSG (1 mM, Invi-
old cycle. The blank controls for these experiments are the trogen, USA), and then further incubated for 30 min.
primers alone without the template cDNA. After 30 min incubation and subsequent 980 nm laser
irradiation (320 mW/cm2; 30 min), the cells were then
trypsinized, aspirated, and suspended in a 1 ml PBS, and
2.15 | Cytotoxicity assays further proceeded to analysis using flow cytometry.

To the 24-well plate, NCI-H23 cells-containing solution


(2.0  104 cells/ml) were added and then incubated 2.18 | In vitro apoptosis detection using
with different amounts of anti-EGFR-UCNPs. The cell Annexin V
solution was incubated at 37 C for 24 h. NCI-H23 cells
were exposed to a 980 nm laser (320 mW/cm2; 30 min) NCI-H23 cells were cultured in a 6-well plate with the
in order to determine the photo-toxicities of anti- cell number of 2  105 cells per well. After 24 hr of incu-
EGFR-UCNPs. Then the cells were incubated for 12 h, bation, different amounts of siRNA-anti-EGFR-UCNPs
followed by addition of 50 μl of a MTT reagent were added and incubated for another 24 hr. After 24 hr,
(0.5 mg/ml). The cells were further incubated for the cells were washed with the PBS, followed by addition
another 4 h. The upper layer was discarded, and 1 ml of serum medium and incubation for additional 24 hr.
of dimethyl sulfoxide (DMSO) was added to each well. The cells were then subjected to photo irradiation with
After that the solution was centrifuged at 13000 rpm, 980 nm laser (320 mW/cm2, 30 min), trypsinized, and
followed by ELISA measurement of optical density at suspended in 200 μl of 1 binding buffer. Then FITC-
570 nm. The optical absorbances were converted to cell Annexin-V (5 μl) antibody (BD Biosciences, USA) and
viabilities based on (absorbance vs. cell numbers) stan- propidium iodide PI (5 μl) were fed to the cell solution
dard curve. and incubated for 5 min and resuspended in fresh PBS
(1 ml), followed by analysis using flow cytometry.

2.16 | ROS experiments


2.19 | In vivo tumor growth study
NCI-H23 cancer cells were seeded in 6-well plates with
a density of 2  105 cells per well. After 24 hr of incuba- The BAlB/c male nude mice were purchased from the
tion, anti-EGFR-UCNPs and siRNA-anti-EGFR-UCNPs National Laboratory Animal Center, Taiwan. The mice
were added to the cancer cells. The cell solution was were raised according to the protocol approved by the
further incubated for 24 hr, followed by photo- institutional animal care and use committee (IACUC) of
irradiation (980 nm, 320 mW/cm2, 30 min). Then, a National Tsing Hua University. Thirty Mice were divided
20 ,70 -dichlorofluorescien diacetate (DCFH-DA) solution into 8 groups on day 0, and used in the in vivo tumor
(5 μM in cell culture medium) was used to replace growth study. Within them, 2 groups (siRNA and anti-
the cell culture medium immediately. DCFH-DA EGFR-UCNPs injected) were treated without photo-
is nonfluorescent and converts to fluorescent 20 ,70 - irradiation and other 6 groups were irradiated by 980 nm
dichlorofluorescien diacetate (DCF) upon reaction with laser (320 mW/cm2; 30 min), once every day. During irra-
the highly active oxygen species, which were generated diation, anti-EGFR-UCNPs (15 mg/kg) and siRNA-anti-
upon photo-irradiation of UCNPs. The cell solution was EGFR-UCNPs (15 mg/kg) were injected thrice (day
further incubated for 30 min at 37 C. Cells were then 1, day 5, and day 9) intravenously. One group of mice
measured using flow cytometry with the FITC channel. was injected with anticancer drug (doxorubicin) with
50 mM NaN3 solution was used for the quenching another group as control without injection of doxorubi-
experiments. cin. Every day, the tumor volumes and body weights
6 KALLURU ET AL.

were measured. The tumor volume (mm3) was calculated


as d2  D/2, where d is the shortest diameter of the
tumor and D is the longest diameter of the tumor. The
relative tumor volume (V/Vo) is calculated by dividing
the volume of the tumor (V) measured on that day to that
of the initial tumor volume (Vo) measured at day 0.

2.20 | HE and caspase 3 staining

Tumor, liver, and spleen were dissected and then sub-


jected to the HE (hematoxylin and eosin) staining. For
caspase-3 staining, only tumor was stained. For tissue
sectioning, 10 μm slice depth was used.

3 | R ES U L T S A N D D I S C U S S I O N S C H E M E 1 Schematic representation for the synthesis of


siRNA-anti-EGFR-UCNPs to facilitate the UCNPs-mediated
3.1 | Synthesis, characterization, and photodynamic therapy combined with SOD1 gene silencing
upconversion luminescence property of
UCNPs
(2H11/2 – 4I15/2), 550 nm (4S3/2 – 4I15/2), and 660 nm (4F9/2
The UCNPs (NaYF4: Yb(30%) and Er(2%)) used in the – 4I15/2), respectively. Since upconverted emission is a
current work were prepared according to the literature two-photon process, both the upconversion emission
procedures.[48] Further, the as-synthesized UCNPs were intensity and quantum yield will become higher at
processed by ligand exchange with the citrate ligands to higher incident laser power intensities.[41] Previously, it
render water dispersibility. The structure of UCNPs was was reported that 45 nm sized NaYF4:Yb3+/Tm3+ hex-
characterized using powder x-ray diffraction (see agonal nanocrystals exhibit an upconverted lumines-
Figure S1(a)). The energy dispersive X-ray analysis of cence quantum yield of ~0.032 upon 980 nm light
UCNPs reveals the presence of Y, Yb, and Er, respectively excitation with a very high laser power intensity of
(Figure S1(b)). To facilitate the combination of UCNPs- 78 W/cm2. Notice that in our system, large upconverted
mediated photodynamic therapy and gene silencing in emission signals were observed upon exceptionally low
treating multidrug resistant lung cancer cells, anti- intensity (25 ~ 150 mW/cm2) of 980 nm NIR excitation
epidermal growth factor receptor (anti-EGFR) was (Figure 1c), of which the incident laser power is 1 ~ 3
chemically linked onto the surface of citrate capped orders lower than the previously reported threshold
UCNPs via EDC amine-acid coupling process (see laser intensity of 1 ~ 1,000 W/cm2 required to observe
Scheme 1).[49,50] The transmission electron microscopy upconversion emission signals.[51] The upconversion
(TEM) images of citrate-capped UCNPs exhibits an aver- emission quantum yields of the as-synthesized and
age particle size of 10 ~ 15 nm. The high resolution TEM citrate-capped UCNPs upon low intensity (0.32 W/cm2)
image in the inset reveals a lattice d-spacing value of 980 nm laser light excitation are determined to be
0.292 nm (see Figure 1a). Dynamic light scattering (DLS) 0.0005% and 0.0002%, respectively (see Figures S4 and
measurements of citrate-capped UCNPs also reveals an S5).[46,52] Although the upconversion luminescence
average particle size of ~10 nm with a polydispersibility quantum yields are very low under ultra-low laser
index (PDI) value of 0.08 (see Figure S2). The successful power excitation, the upconversion emission intensities,
chemical conjugation of anti-EGFR onto the surface of however, are still large enough to be detected and visu-
UCNPs was confirmed by the UV–vis absorption spec- alized (see Figure 1c).
trum, which reveals an absorption peak at ~280 nm, a The UV–vis–NIR spectrum in Figure 2a shows that
characteristic absorption of the anti-EGFR (Figure 1b). citrate-capped UCNPs exhibit flat absorption extending
The citrate capping and subsequent anti-EGFR linking up to the NIR region (1,100 nm) along with a small peak
on the surface of the UCNPs were also confirmed by at 976 nm, which is characteristic for the absorption of
FTIR spectroscopic analysis (see Figure S3).[50] Upon Yb(III) ions.[53] The molar extinction coefficient for
excitation of 980 nm light, citrate-capped UCNPs reveals citrate-capped UCNPs was determined for the first time
three upconversion emission peaks centered at 525 nm to be ~2.0 x 107 M1/cm at 980 nm, which is 3 ~ 4 orders
KALLURU ET AL. 7

F I G U R E 1 Characterization of UCNPs. (a) Transmission electron microscopy (TEM) image of bare UCNPs. The inset represents the
high-resolution TEM image of citrate-capped UCNPs. (b) UV–vis absorption spectra for citrate-capped UCNPs, anti-EGFR alone, and
siRNA-anti-EGFR-UCNPs, respectively. The green and red dashed arrows indicate their respective absorption peak at 280 nm for anti-EGFR
alone and siRNA-anti-EGFR-UCNPs. (c) Upconverted emission spectra measured using a 980 nm laser with different power intensities. Inset
is the digital luminescence photograph of citrate-capped UCNPs

F I G U R E 2 Optical properties of UCNPs. (a) UV–vis–NIR absorption spectrum (black solid line) and excitation spectrum (black dashed
line) for singlet oxygen phosphorescence of citrate-capped UCNPs (λem = 1,261 nm). (b) Singlet oxygen phosphorescence emission spectra
sensitized by citrate capped UCNPs, in the absence of organic photosensitizers, at 980 and 1,008 nm excitation wavelengths, respectively

higher than the conventional organic dyes, but 1 ~ 2 singlet O2 by itself upon 980 nm light excitation in the
orders smaller than those for spherical gold nanoparticles absence of organic photosensitizers. The excitation spec-
and short aspect ratio gold nanorods.[27,54] In addition to trum for singlet O2 phosphorescence from UCNPs reveals
the upconverted emission, amazingly we also found that three characteristic peaks centered at 947, 980, and
the citrate-capped UCNPs can sensitize formation of 1,008 nm (see dashed lines in Figure 2a). Interestingly,
8 KALLURU ET AL.

singlet O2 can only be formed when excited by NIR light 3.2 | In vitro combined UCNPs-mediated
(875 ~ 1,100 nm), but not by visible and short NIR light. PDT and gene silencing
Upon excitation at 980 nm and 1,008 nm, very clear-cut
singlet O2 phosphorescence emission signals were To evaluate whether combination of UCNP-mediated
detected, with an emission maximum at ~1,261 nm PDT and gene silencing can kill multidrug resistant non-
(Figure 2b). The quantum yield for the sensitization of small lung cancer cells, anti-EGFR surface-modified
singlet O2 by UCNPs was determined to be 0.353 at UCNPs were fed to the NCI-H23 cells, and the cellular
980 nm (at an incident laser intensity of 320 mW/cm2, uptake was monitored. The confocal optical images in
see Data S1 for more details). The generation of singlet Figure S7 and S8 clearly show that the anti-EGFR
O2 by photo excitation of UCNPs was also confirmed surface-modified UCNPs were successfully uptaken by
using a chemical trapping agent, 1,3-diphenyl iso- the NCI-H23 cells, as evidenced by the observation of
benzofuran (DPBF). DPBF reacts with singlet O2 via upconverted red fluorescence emission image upon
1,4-cycloaddition to form an endoperoxide product, 980 nm laser light excitation. In contrast, citrate-UCNPs
which leads to a decrease in the absorption intensity at internalized NCI-H23 cells as well as the control (without
410 nm.[55] As shown in Figure S6(a), the 410 nm absorp- any pre-treatment) did not show any noticeable
tion intensities of the solution containing UCNPs and upconverted emission. Further, we have loaded SOD1
DPBF decreases gradually upon prolonged 980 nm siRNA together with the anti-EGFR-UCNPs, aiming at
photo-irradiation, whereas no or negligible change was gene therapy to kill non-small lung cancer cells. The
observed at 410 nm from two control solutions: (a) a zeta-potential measurements in Figure 3a confirm the
DPBF-containing solution (without UCNPs) under complexation, via electrostatic interactions, of poly-
photo-irradiation, and (b) a DPBF-UCNPs solution in the anionic siRNA onto the surface of cationic anti-EGFR-
dark. In addition to the observation of singlet O2 phos- coated UCNPs. The gene silencing efficiencies of SOD1
phorescence emission and DPBF experiments upon using siRNA-anti-EGFR coated UCNPs as nanocarriers
980 nm light excitation of citrate-UCNPs, we have also were evaluated using quantitative polymerase chain reac-
detected 1O2 using electron paramagnetic resonance tion (qPCR) in NCI-H23 cells under serum free condi-
(EPR) spectroscopy. To detect 1O2 using EPR, we have tions (see Figure 3b and c). Overall, ~70% gene silencing
used 2,2,6,6,-tetramethyl-4-piperidone (TEMP) as a 1O2 efficiency of SOD1 was achieved in the NCI-H23 cells
spin-trapping reagent. The EPR signal for the photoex- after pre-treatment with siRNA-anti-EGFR-coated
cited citrate-UCNPs dispersion clearly displayed a charac- UCNPs (at 100 nM siRNA concentration). Therefore, it is
teristic 1:1:1 triplet due to the formation of a TEMP-1O2 anticipated that upon silencing of SOD1 gene, the
spin adduct. Further, the TEMP-1O2 spin adduct EPR sig- siRNA-anti-EGFR-UCNPs might be able to mediate pho-
nal was remarkably suppressed in the presence of a spe- todynamic therapeutic effects, upon NIR light irradiation,
cific singlet oxygen scavenger, L-histidine (see Figure S6 on killing the NC1H23 lung cancer cells effectively. To
(b)). Overall, it is very clear that upon photo-excitation prove such a possibility and to evaluate the cancer cell-
using 980 nm light, citrate-UCNPs alone in the absence killing efficiency, NCI-H23 cells were incubated with dif-
of additional organic photosensitizers can effectively sen- ferent concentrations of anti-EGFR-UCNPs and siRNA-
sitize the formation of singlet oxygen. All these results anti-EGFR-UCNPs and irradiated with 980 nm laser. As
clearly point to the same conclusion that citrate-capped shown in Figure 3d, neither anti-EGFR-UCNPs nor
UCNPs alone in the absence of organic photosensitizers siRNA-anti-EGFR-UCNPs induce any noticeable cellular
can sensitize formation of singlet O2 as well as deaths in the dark. However, upon exposure to 980 nm
upconverted emission upon 980 nm light excitation. The laser (320 mW/cm2; 30 min), the percentage of cellular
photo-sensitization of singlet O2 from UCNPs was based deaths from the siRNA-anti-EGFR-UCNPs group
on single photon excitation and subsequent energy trans- increases dramatically, which is ~3.8 fold higher than
fer process from the excited state of UCNPs to molecular that observed from the anti-EGFR-UCNPs group without
O2. To have energy transfer occur to generate singlet O2, silencing gene. In contrast, siRNA (100 nM concentra-
molecular O2 most probably adsorbs on the surface of the tion) alone did not induce any significant cellular deaths
UCNPs. In other words, the singlet excited state energy both in the dark and siRNA (100 nM concentration)
of UCNPs was estimated to be ~1.26 eV (i.e., E = 1,239/ upon photo irradiation condition. To provide deeper
λex = 1239/980 nm = 1.26 eV or 29.17 kcal/mol), which insights into the cellular deaths by anti-EGFR-UCNPs
is much larger than the formation energy of singlet O2, and siRNA-anti-EGFR-coated UCNPs internalized NCI-
0.97 eV (i.e., 22.5 kcal/mol). Overall, an energy transfer H23 cells using 980 nm laser irradiation, several path-
mechanism from UCNPs to molecular O2 can lead to the ways were investigated to understand the mechanisms
generation of singlet oxygen. which lead to high cellular deaths. From Figure 4a, it
KALLURU ET AL. 9

F I G U R E 3 Combined in vitro UCNPs-mediated photodynamic therapy and gene silencing. (a) Zeta-potential measurements for citrate
UCNPs, anti-EGFR-UCNPs and various concentrations of siRNA-anti-EGFR-UCNPs solutions. (b) Percentage of gene silencing efficiencies
measured using RT-qPCR for SOD1 gene at various concentrations of siRNA. (c) Western blotting result for the SOD1 gene at various siRNA
concentrations with β-Actin as an internal control. (d) Percentages of cellular deaths of siRNA, anti-EGFR-UCNPs and siRNA-anti-EGFR
UCNPs internalized NCI-H23 cells in the dark and under photo irradiation (980 nm, 320 mW/cm2; 30 min) conditions at 37 C

clearly reveals that upon 980 nm laser irradiation the origin for these elevated levels of ROS were never
(320 mW/cm2; 30 min), the ROS levels for anti-EGFR addressed in these previous reports.[55–57] Our results
UCNPs and siRNA-anti-EGFR UCNPs treated NCI-H23 can help rationalize these previous observations, which
cells were ~2.0 and ~2.6-fold, respectively, higher than are, in fact, due to the direct sensitization of singlet oxy-
that for the dark control group. In contrast, in the gen by photochemically excited UCNPs. The fluores-
absence of photo irradiation the ROS levels for both cence intensity levels of singlet O2 sensor green (SOSG)
anti-EGFR modified UCNPs and siRNA-anti-EGFR in Figure S10 exhibit ~2.2-fold higher, as compared to
UCNPs are about the same as that of the control cells that in the dark, upon 980 nm laser irradiation for both
(see Figure S9 for confocal images). To further confirm anti-EGFR-UCNPs and siRNA-anti-EGFR-coated
the role of singlet O2 on cellular deaths, NCI-H23 cells UCNPs systems. From Figure 4c, we also observed that
were pre-treated with a singlet O2-specific quencher, the amounts of apoptotic and necrotic cells generated
that is, sodium azide (NaN3).[27–31] The ROS levels were from siRNA-anti-EGFR-coated UCNPs-internalized
significantly suppressed by addition of sodium azide for NCI-H23 cells upon 980 nm light irradiation are far
both anti-EGFR-UCNPs and siRNA-anti-EGFR-UCNPs higher than those for anti-EGFR-UCNPs treated group
groups in the case of 980 nm photo irradiation (see in the dark. Overall, our in vitro experiments clearly
Figure 4b). In a few previous studies, significant show that a combination of UCNPs-mediated PDT and
amounts of ROS levels and its subsequent cellular gene silencing can conquer the MDR problem and exerts
deaths were observed for the UCNPs alone when effective killing of the NCI-H23 lung cancer cells in
exposed to 980 nm NIR light irradiation.[55–57] However, in vitro experiments.
10 KALLURU ET AL.

F I G U R E 4 In vitro ROS generation and apoptosis. (a) ROS generation monitored by the mean fluorescence intensities of 20 ,70 -
dichlorofluorescein (DCF) using flow cytometry for anti-EGFR-UCNPs and siRNA-anti-EGFR-UCNPs internalized NCI-H23 cells followed
by photo irradiation. (b) ROS levels observed after 50 mM NaN3 pretreatment. (c) Percentages of apoptotic and necrotic cell populations
obtained by the NCI-H23 cells treated with anti-EGFR-UCNPs and siRNA-anti-EGFR-UCNPs in the dark and under photo irradiation
(980 nm, 320 mW/cm2; 30 min) conditions

3.3 | In vivo combination of UCNPs- Although less than 5% of siRNA-anti-EGFR UCNPs


mediated PDT and gene silencing on nanoconjugates reach the tumor site, the amount is
tumor destruction enough to sensitize formation of singlet oxygen efficiently
and achieve complete destruction of lung tumors under
Based on the effective killing of the intrinsically our experimental conditions (vide infra). At 24 hr after
multidrug resistant NCI-H23 lung cancer cells in the intravenous injection of siRNA alone or anti-EGFR-
in vitro experiments, we took one step further to deter- UCNPs or siRNA-anti-EGFR-UCNPs, the NCI-H23
mine, as a proof-of-concept study, the effectiveness of the tumor bearing mice was subjected to 980 nm NIR light
combined UCNPs-mediated PDT and gene silencing on irradiation. As shown in Figure 5a, the mice treated with
in in-vivo NCI-H23 non-small cell lung tumor destruc- siRNA-anti-EGFR-UCNPs and exposed to 980 nm laser
tion in a BALB/c nude mice model. The NCI-H23 tumors have complete destruction of tumors at day 20th, which
were implanted subcutaneously on the leg muscle region, is in sharp contrast to the much larger tumor sizes of 2.0
and allowed to grow until it reaches a diameter of and 26-fold (relative to the initial tumor size) for anti-
~5 mm. The siRNA-anti-EGFR-UCNPs (15 mg/kg) EGFR-UCNPs (without silencing gene) and siRNA alone
nanoconjugates were then injected intravenously. At (without UCNPs) plus exposure to 980 nm laser light.
24 hr after iv injection, the distribution of UCNPs in vari- The sizes (regions indicated in the dashed white circles)
ous organs, including tumor, liver, spleen, and kidney, of the NCI-H23 tumors on mice treated with different
were examined using inductively coupled plasma mass conditions at day 20th were presented in Figure 5b. It is
spectrometry (ICP-MS) analysis (see Figure S11). also interesting to note that the tumor site of the mice
KALLURU ET AL. 11

F I G U R E 5 Combined in vivo UCNPs-mediated photodynamic therapy and gene silencing. (a) Tumor growth curves of different groups
of tumor-bearing mice after treatment. The tumor volumes were normalized to the initial size at day 0. Asterisks indicate statistically
significant differences (p < .001) between all the pairs. (b) Representative mice images showing the size of the tumors indicated by dashed
white circles at day 20 after treatment with siRNA alone or anti-EGFR-UCNPs or siRNA-anti-EGFR-UCNPs exposure to 980 nm
(320 mW/cm2; 30 min) light, whereas the blank controls receive no any treatment. The representative mice images showing the scar
formation at day 20th and day 30th, followed by a new skin formation at day 40th for siRNA-anti-EGFR-UCNPs treated mice and exposure
to 980 nm light. (c) In vivo fluorescence microscopy images for the caspase 3 staining of the tumor tissue dissected from various treatment
groups

treated with siRNA-anti-EGFR-UCNPs and exposure to with siRNA-anti-EGFR-UCNPs plus exposure to 980 nm
980 nm laser light forms a scar at day 20th, which got laser is longer than 40 days (4 mice completely survive
peeled off later and a new skin was then gradually devel- out of 6 mice monitored at day 40th in the combined
oped at day 40th (see Figure 5b). The results unambigu- UCNPs-mediated PDT and gene silencing with photo
ously demonstrate that the multidrug resistant NCI-H23 irradiation group), which is much longer than the mice
non-small cell lung tumor could be destroyed completely treated with anti-EGFR-UCNPs and exposure to 980 nm
by the combined UCNPs-mediated PDT and SOD1 gene laser group (28 days), doxorubicin treated mice group
silencing. Besides the tumor growth data presented in the (15 days) as well as PBS-treated and photo irradiated
Figure 5a and b, other measurements, such as, caspase mice group (17 days), respectively (see Figure 6a). During
activity and mice survival rate, are also consistent with the therapy period, there are no noticeable changes in
each other. The caspase activity of tumor tissues dissected the body weights for all the mice groups (see Figure 6b).
from those mice treated with siRNA-anti-EGFR-UCNPs Hematoxylin & Eosin (HE) staining in Figure S12 from
and exposure to 980 nm laser exhibit higher expression of siRNA alone-treated mice followed by photo irradiation.
caspase 3 than those treated with anti-EGFR-UCNPs or In contrast, severe necrosis was observed for the tumor
siRNA alone and exposure to photo irradiation (see tissues dissected from the siRNA-anti-EGFR-UCNPs or
Figure 5c). The average half-life span for the mice treated anti-EGFR-UCNPs treated mice and exposure to 980 nm
12 KALLURU ET AL.

F I G U R E 6 (a) Survival rates of the


mice bearing NCIH23 lung tumors after
various treatments monitored for
40 days. (b) Body weight measurements
of the mice bearing NCIH23 lung
tumors for various treatment groups

light. Overall, our in vivo results, consistent with the models. The average lifespan (>40 days) of tumor bearing
in vitro cellular data, clearly demonstrate a successful mice treated with combined UCNPs-mediated PDT and
example of complete destruction of multidrug resistant gene silencing is ~2.5 times longer than that (15 days) of
lung tumors by combination of unprecedented NIR light anticancer drug (doxorubicin)-treated group. To the best
activatable UCNPs-mediated PDT and SOD1 gene of our knowledge, this is the first successful literature
silencing. example of achieving complete destruction of multidrug
resistant cancers. Overall, our work has pointed a new
direction for effective therapeutic design to completely
4 | C ON C L U S I ON S conquer multidrug resistance problems commonly associ-
ated in many cancers.
In summary, we have presented a successful therapeutic
design, via combination of UCNPs-based PDT and gene ACKNOWLEDGMENTS
silencing, to conquer the tumor multidrug resistance The authors are grateful to prof. Hsing-Wen Sung,
problem. As a proof-of-concept experiment, we have Department of Chemical Engineering, National Tsing
demonstrated that multidrug resistant NCI-H23 lung Hua University, Hsinchu, Taiwan for providing the RT-
tumor in a mice model can be completely destructed by qPCR instrumentation facility, and also to the financial
an unprecedented UCNPs-mediated photodynamic ther- support from Frontier Research Center on Fundamental
apy combined with efficient SOD1 gene silencing (~70%) and Applied Sciences of Matters, National Tsing Hua
using ultra-low doses (320 mW/cm2) of NIR light University, and Ministry of Science and Technology,
(980 nm). The multi-functional UCNPs act as an unique Taiwan.
theranostic reagent to provide the following functions,
including (a) in vivo tumor fluorescence imaging probe ORCID
(via its NIR-activatable upconverted emission property), Kuo Chu Hwang https://orcid.org/0000-0003-1814-9869
where we report for the first time that upconversion
emission can be observed under an exceptionally low RE FER EN CES
intensity of 980 nm (25 ~ 50 mW/cm2) light excitation; [1] M. M. Gottesman, T. Fojo, S. E. Bates, Nat. Rev. Cancer 2002,
(b) ROS can be generated directly by NIR light photo- 2, 48.
excitation of UCNPs in the absence of additional organic [2] C. F. Higgins, Nature 2007, 446, 749.
[3] C. Holohan, S. van Schaeybroeck, D. B. Longley, P. G.
photosensitizers, and (c) combination of NIR-I PDT with
Johnston, Nat. Rev. Cancer 2013, 13, 714.
SOD1 gene silencing for eradication of multidrug resis- [4] G. Szakacs, J. K. Paterson, J. A. Ludwig, C. Booth-Genthe,
tant lung tumor. M. M. Gottesman, Nat. Rev. Drug Discov. 2006, 5, 219.
In the in vitro cellular experiments, upon photo exci- [5] G. V. Scagliotti, S. Novello, G. Selvaggi, Ann. Oncol. 1999, 10-
tation of 980 nm light, significantly higher amounts of (Suppl 5), S83.
cellular deaths were observed from the siRNA-anti-EGFR [6] M. Shanmugam, N. Kuthala, R. Vankayala, C.-S. Chiang, X.
UCNPs treated NCI-H23 cells, as compared to that Kong, K. C. Hwang, ACS Nano 2021, 15, 14404.
[7] G. R. Bertolini, L. Perego, P. Tortoreto, M. Fontanella, E.
treated with anti-EGFR UCNPs alone without silencing
Gatti, L. Pratesi, G. Fabbri, A. Andriani, F. Tinelli, S. Roz, E.
gene (79% vs. 33% cellular deaths at 50 μg/ml concentra-
Caserini, R. Lo Vullo, S. Camerini, T. Mariani, L. Delia, D.
tions). In the in vivo experiments, combination of the Calabrò, E. Pastorino, U. Sozzi, Proc. Natl. Acad. Sci. U. S. A.
unprecedented UCNPs-mediated PDT and gene silencing 2009, 106, 7779.
was demonstrated to completely destruct the multidrug [8] X. Zhu, Y. Xu, L. M. Solis, W. Tao, L. Wang, C. Behrens, X.
resistant non-small cell NCI-H23 lung tumors in mice Xu, L. Zhao, D. Liu, J. Wu, N. Zhang, O. C. Wistuba II., B. R.
KALLURU ET AL. 13

Farokhzad, J. S. Zetter, Proc. Natl. Acad. Sci. U. S. A. 2015, [38] N. Kuthala, R. Vankayala, C.-S. Chiang, K. C. Hwang, Adv.
112, 7779. Funct. Mater. 2020, 30(36), 2002940.
[9] Y. Qian, M. Qiu, Q. Wu, Y. Tian, Y. Zhang, N. Gu, S. Li, L. Xu, [39] X. Kong, R. Cheng, J. Wang, Y. Fang, K. C. Hwang, Nano
R. Yin, Sci. Rep. 2014, 4, 7490. Today 2021, 36, 101004.
[10] H. Zhao, F. Yang, W. Shen, Y. Wang, X. Li, J. You, Q. Zhou, [40] M. Shanmugam, N. Kuthala, R. Vankayala, C.-S. Chiang, X.
Thorac. Cancer 2015, 6, 133. Kong, K. C. Hwang, ACS Nano 2021, 15(9), 14404.
[11] A. Gulzar, J. Xu, D. Yang, L. Xu, F. He, S. Gai, P. Yang, Dal- [41] T. Yokota, M. Miyagishi, T. Hino, R. Matsumura, A. Tasinato, M.
ton. Trans. 2018, 47, 3931. Urushitani, R. V. Rao, R. Takahashi, D. E. Bredesen, K. Taira, H.
[12] A. Gulzar, Z. Wang, F. He, D. Yang, F. Zhang, S. Gai, P. Yang, Mizusawa, Biochem. Biophys. Res. Commun. 2004, 314, 283.
Inorg. Chem. 2020, 59, 4909. [42] C. Raoul, T. Abbas-Terki, J. C. Bensadoun, S. Guillot, G. Haase,
[13] Q. Cai, J. Xu, D. Yang, Y. Dai, G. Yang, C. Zhong, S. Gai, F. J. Szulc, C. E. Henderson, P. Aebischer, Nat. Med. 2005, 11, 423.
He, P. Yang, J. Mater. Chem. B. 2018, 6, 8148. [43] R. Somwar, H. Erdjument-Bromage, E. Larsson, D. Shum,
[14] D. W. Felsher, Nat. Rev. Cancer 2003, 3, 375. W. W. Lockwood, G. Yang, C. Sander, O. Ouerfelli, P. J.
[15] A. P. Castano, P. Mroz, M. R. Hamblin, Nat. Rev. Cancer 2006, 6, 535. Tempst, H. Djaballah, H. E. Varmus, Proc. Natl. Acad. Sci.
[16] R. Vankayala, K. C. Hwang, Adv. Mater. 2018, 30, 1706320. U. S. A. 2011, 108, 16375.
[17] A. Gulzar, J. Xu, L. Xu, P. Yang, F. He, D. Yang, G. An, M. B. [44] S. S. Kim, C. Ye, P. Kumar, I. Chiu, S. Subramanya, H. Wu, P.
Ansari, Dalton Trans. 2018, 47, 3921. Shankar, N. Manjunath, Mol. Ther. 2010, 18, 993.
[18] M.-H. Chan, S.-P. Chen, C.-W. Chen, Y.-C. Chan, R. J. Lin, [45] D. Yang, J. Xu, G. Yang, Y. Zhou, H. Ji, H. Bi, S. Gai, F. He, P.
D. P. Tsai, M. Hsiao, R.-J. Chung, X. Chen, R.-S. Liu, J. Phys. Yang, Chem. Eng. J. 2018, 344, 363.
Chem. C 2018, 122, 2402. [46] S. Pattingre, A. Tassa, X. Qu, R. Garuti, X. H. Liang, N.
[19] X. Wu, Y. Zhang, Z. Wang, J. Wu, R. Yan, C. Guo, Y. Jin, Mizushima, M. Packer, M. D. Schneider, B. Levine, Cell 2005,
A. C. S. Appl, Bio. Mater. 2020, 3, 5813. 122, 927.
[20] B. Güleryüz, U. Ünal, M. Gülsoy, Photodiagn. Photodynam. [47] X. Wu, Y. Zhang, Z. Wang, J. Wu, R. Yan, C. Guo, Y. Jin, ACS
Therapy 2021, 36, 102616. Appl. Bio Mater. 2020, 3, 5813.
[21] P. Kowalik, I. Kami nska, K. Fronc, A. Borodziuk, M. Duda, T. [48] Q. Liu, W. Feng, T. Yang, T. Yi, F. Li, Nat. Protoc. 2013, 8, 2033.
Wojciechowski, K. Sobczak, D. Kalinowska, M. T. Klepka, B. [49] X. H. Ivan, H. El-Sayed, M. A. El-Sayed, Nano Lett. 2005,
Sikora, Nanotechnology 2021, 32, 475101. 5, 829.
[22] N. M. Idris, M. K. Gnanasammandhan, J. Zhang, P. C. Ho, R. [50] X. Hun, Z. Zhang, Spectrochim. Acta A Mol. Biomol. Spectrosc.
Mahendran, Y. Zhang, Nat. Med. 2012, 18, 1580. 2009, 74, 410.
[23] P. S. Can, T. Xu, H. Liu, X. Wu, M. E. Messing, L. R. [51] S. Han, R. Deng, X. Xie, X. Liu, Angew Chem. 2014, 53, 11702.
Wallenberg, S. Andersson-Engels, ACS Nano 2012, 6, 4788. [52] G. S. Chen, J. Ohulchanskyy, T. Y. Patel, N. J. Kutikov, A. Li,
[24] X. Liu, I. Que, X. Kong, Y. Zhang, L. Tu, Y. Chang, T. T. Wang, Z. Song, J. Pandey, R. K. Ågren, H. Prasad, P. N. Han, ACS
A. Chan, C. W. Lowik, H. Zhang, Nanoscale. 2015, 7, 14914. Nano 2012, 6, 8280.
[25] C. Wang, L. Cheng, Y. Liu, X. Wang, X. Ma, Z. Deng, Y. Li, Z. [53] W. Zou, C. Visser, J. A. Maduro, M. S. Pshenichnikov, J. C.
Liu, Adv. Funct. Mat. 2013, 23, 3077. Hummelen, Nature Photonic. 2012, 6, 560.
[26] D. Y. Sisi Cui, Y. Chen, Y. Di, H. Chen, Y. Ma, S. Achilefu, Y. [54] X. Liu, M. Atwater, J. Wang, Q. Huo, Colloids Surf. B Bio-
Gu, ACS Nano. 2013, 7, 676. interfaces 2007, 58, 3.
[27] R. Vankayala, Y. K. Huang, P. Kalluru, C. S. Chiang, K. C. [55] R. Vankayala, C. L. Kuo, A. Sagadevan, P. H. Chen, C. S.
Hwang, Small 2014, 10, 1612. Chiang, K. C. Hwang, J. Mater. Chem. B 2013, 1, 4379.
[28] R. Vankayala, C. C. Lin, P. Kalluru, C. S. Chiang, K. C. [56] Y. I. Park, H. M. Kim, J. H. Kim, K. C. Moon, B. Yoo, K. T.
Hwang, Biomaterials 2014, 35, 5527. Lee, N. Lee, Y. Choi, W. Park, D. Ling, K. Na, W. K. Moon,
[29] P. Vijayaraghavan, C. H. Liu, R. Vankayala, C. S. Chiang, S. H. Choi, H. S. Park, S. Y. Yoon, Y. D. Suh, S. H. Lee, T.
K. C. Hwang, Adv. Mater. 2014, 26, 6689. Hyeon, Adv. Mater. 2012, 24, 5755.
[30] P. Vijayaraghavan, R. Vankayala, C. S. Chiang, H. W. Sung, [57] T. Wei, C. Chen, J. Liu, C. Liu, P. Posocco, X. Liu, Q. Cheng,
K. C. Hwang, Biomaterials 2015, 62, 13. S. Huo, Z. Liang, M. Fermeglia, S. Pricl, X. J. Liang, P. Rocchi,
[31] P. Kalluru, R. Vankayala, C. S. Chiang, K. C. Hwang, Angew. L. Peng, Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 2978.
Chem. 2013, 52, 12332.
[32] R. Vankayala, C. L. Kuo, K. Nuthalapati, C.-S. Chiang, K. C.
Hwang, Adv. Funct. Mater. 2015, 25, 5934. SU PP O R TI N G I N F O RMA TI O N
[33] P. Kalluru, R. Vankayala, C.-S. Chiang, K. C. Hwang, Biomate- Additional supporting information may be found in the
rials 2016, 95, 1. online version of the article at the publisher's website.
[34] P. Vijayaraghavan, C.-S. Chiang, H. K. Chiang, M.-L. Li, K. C.
Hwang, Adv. Mater. Technol. 2016, 1, 1600107.
[35] P. Vijayaraghavan, C.-H. Liu, K. C. Hwang, A. C. S. Appl, How to cite this article: P. Kalluru, M.
Mater. Interfaces 2016, 8, 23909. Shanmugam, R. Vankayala, C.-S. Chiang, K. C.
[36] P. Kalluru, R. Vankayala, C.-S. Chiang, K. C. Hwang, Adv. Hwang, J. Chin. Chem. Soc. 2022, 1. https://doi.org/
Funct. Mater. 2016, 26, 7908. 10.1002/jccs.202200001
[37] N. Kuthala, R. Vankayala, Y.-N. Li, C.-S. Chiang, K. C.
Hwang, Adv. Mater. 2017, 29, 1700850.

You might also like