Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Acta Biomaterialia 135 (2021) 164–178

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actbio

Full length article

Intelligent and spatiotemporal drug release based on multifunctional


nanoparticle-integrated dissolving microneedle system for synergetic
chemo-photothermal therapy to eradicate melanoma
Yiting Zhao a,1, Yixian Zhou a,1, Dan Yang b, Xinyi Gao b, Ting Wen a, Jintao Fu a, Xinguo Wen c,
Guilan Quan b,∗, Xin Pan a,∗, Chuanbin Wu b
a
School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
b
College of Pharmacy, Jinan University, Guangzhou 510632, China
c
Guangzhou Neworld Micnanobio Pharmatech Co., Ltd, Guangzhou 510670, China

a r t i c l e i n f o a b s t r a c t

Article history: Cutaneous melanoma is one of the most common malignant skin cancer with high lethality. Chemother-
Received 24 May 2021 apy and photothermal therapy are important and extensively studied treatment modalities for melanoma.
Revised 3 September 2021
However, these therapies still face some challenges, which severely restrict their further applications, such
Accepted 7 September 2021
as unsatisfactory efficacy of monotherapy, nonspecific uptake and release during drug delivery, and un-
Available online 13 September 2021
expected adverse effects from system administration. Recently, the strategies of collaboration, functional
Keywords: modification, stimuli-responsive design, and topical administration all show great prospect for solving
Cellular uptake above problems. In this research, a multifunctional nanoparticle-integrated dissolving microneedle drug
Specific distribution delivery system was constructed, in which the nanoparticles were prepared based on the framework with
Synergistic therapy the incorporation of photothermal agent (CuS) into Zeolitic imidazolate framework-8 and functionalized
Dissolving microneedles by hyaluronic acid. This system can co-load multi-modal drugs, improve specific uptake and distribu-
Superficial tumor
tion of targeted tumor, deliver drug locally, and release drug intelligently and spatiotemporally, thereby
promising a low-dose administration with high efficiency. The high inhibiting tumor performance and
excellent systematic safety were verified both in vitro and in vivo. Together, this smart design overcame
the drawbacks of monotherapy and conventional system administration. We believe the nanoparticle-
integrated dissolving microneedles will be in prospect of clinical application for more superficial tumors
with further delicate optimization.

Statement of significance

Melanoma is one of the most common skin cancers with high lethality. Extensively studied chemother-
apy and photothermal therapy still face some challenges, such as the limited therapeutic efficacy and the
severe system adverse effects. In order to overcome these drawbacks, the multifunctional nanoparticle-
integrated dissolving microneedles (DMNs) were designed. Especially, the nanoparticles could co-load
multi-modal drugs, improve specific uptake, and release drug intelligently and spatiotemporally. The mi-
croneedles could increase the drug accumulation in tumor, thus achieving excellent therapeutic efficacy
and reducing side effects. This system paved the way to a less invasive, more focused and efficient ther-
apeutic strategy for melanoma therapy.
© 2021 Published by Elsevier Ltd on behalf of Acta Materialia Inc.

1. Introduction cer, resulting in nearly 12 thousand deaths yearly [1,2]. In the con-
text of skin cancer, melanoma is the most life-threatening type
Skin cancer has been one of the ruthless killers for human [2] and responsible for over 75% of deaths [3]. Chemotherapy and
health. In USA, over 10 0,0 0 0 patients are diagnosed with skin can- photothermal therapy (PTT) are important and extensively studied
treatment modalities for melanoma. Chemotherapy directly kills

Corresponding authors. susceptible cancer cells by damaging the chromosome structural
E-mail addresses: quanguilan@jnu.edu.cn (G. Quan), panxin2@mail.sysu.edu.cn integrity and disrupting DNA synthesis [4], and PTT can destroy
(X. Pan). tumor cell membranes and denature proteins via hyperthermia to
1
These authors equally contributed to this work.

https://doi.org/10.1016/j.actbio.2021.09.009
1742-7061/© 2021 Published by Elsevier Ltd on behalf of Acta Materialia Inc.
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

induce tumor cell apoptosis [5–8]. However, the above therapies drophobic chemotherapeutic drug (Camptothecin, CPT) into ZIF-8
still face some challenges, which severely restrict their further ap- framework. Subsequently, the nanoparticles were coated with a
plications. First, the therapeutic efficacy of monotherapy is unde- HA shell, denoted as CPT-CuS-ZIF-8@HA (Scheme 1A). Finally, CPT-
sirable due to tumorous complexity and heterogeneity. The strat- CuS-ZIF-8@HA nanoparticles were loaded into the tips of DMNs
egy focused on combination therapy has attracted considerable at- through molding method [42] for local and synergetic therapy
tention, which can achieve additive and/or synergistic antitumor (Scheme 1B). The successfully synthesized CPT-CuS-ZIF-8@HA
efficacy through pleiotropic and complex mechanisms. For exam- nanoparticles were characterized by scanning electron microscopy
ple, in combination therapy based on PTT and chemotherapy, PTT (SEM), dynamic light scattering (DLS), Fourier transform infrared
can cause cellular damage by hyperthermia, simultaneously the el- ray (FT-IR), powder X-ray diffraction (PXRD), thermogravimetric
evated temperature can increase the blood flow to accelerate drug analysis (TGA), and ultra-violet and visible (UV-Vis) measurement.
delivery into deep-seated tumors, thus achieving synergistic effi- As expected, CPT-CuS-ZIF-8@HA could achieve intelligent and spa-
cacy [9–12]. Second, nonspecific uptake and release during drug tiotemporal control drug release under low pH and near infrared
delivery cause decreased damage for tumor cells. To tackle this ray (NIR) irradiation in vitro. In addition, the results of cellular
problem, intelligent nano-formulation exploitation [13–15], func- uptake suggested HA modification could significantly improve
tionally targeted modification [16,17], and spatiotemporally con- the internalization of nanoparticles. Moreover, in vivo biodistribu-
trolled release design [18,19] are introduced. Third, system admin- tion and pharmacodynamics further confirmed the topical drug
istration remains the most conventional mode for tumor therapy, enrichment and synergetic multi-modal therapy of the DDS. In
while it always results in severe system adverse effects and con- general, the strategic integration of DMNs and multifunctional
siderable drug wastage in local tumor therapy. Therefore, topical nanoparticles exhibited superior advantages in intelligent and
drug delivery may be an effective solution to concentrate drugs in spatiotemporal drug delivery for superficial tumor therapy, and
the tumor site and reduce drugs absorbed into blood circulation, we believe the future designs of diverse hierarchical targeting and
thereby minimizing side effects. stimuli-responsivity could further broaden the therapeutic range.
Metal organic frameworks (MOFs) are a type of porous mate-
rial with uniform and tunable porosity [20–23]. Among them, Ze- 2. Experimental section/methods
olitic imidazolate framework-8 (ZIF-8) is one of the most represen-
tative MOFs built from zinc ions and 2-methylimidazolate (2-Mim), 2.1. Materials
and is regarded as a promising drug carrier in tumor treatment
due to its unique advantages [24–27]. For example, ZIF-8 can load CPT and Copper(II) chloride dehydrate (CuCl2 ·2H2 O) were ac-
multi-modal drugs on account of its unique porous interior. In ad- quired from Shanghai Macklin Biochemical Co., Ltd. (Shanghai,
dition, owing to the pH-responsive degradation of ZIF-8, the ZIF-8 China). HA, Zinc nitrate hexahydrate (Zn(NO3 )2 6·H2 O), 2-Mim, flu-
based nanoparticles can release drugs intelligently in the acid tu- orescein (FL), polyvinyl alcohol (PVA 103), and sodium sulfide
mor microenvironments (pH 5.5-6.0) [28,29]. Simultaneously, the (Na2 S) were supplied by Aladdin Bio-Chem Technology Co., Ltd.
zinc ions and 2-Mim produced by decomposition are nontoxic and (Shanghai, China). Polyvinyl Pyrrolidone (PVP-K30, 40 kDa) and 4 ,
easy to be metabolized and excreted under physiological condi- 6-Diamidino-2-phenylindole (DAPI) were purchased from Beijing
tions, demonstrating its high biocompatibility. Apart from that, due Solarbio Science & Technology Co., Ltd. (Beijing, China). Annexin
to the adjustable surface functionality, ZIF-8 can be modified to V-FITC/Propidium Iodide (PI) was obtained from MultiSciences
improve active targeting, reduce rapid clearance, prolong blood cir- Biotech Co., Ltd. (Hangzhou, China). Calcein-AM/PI kit was supplied
culation, and so on. For instance, hyaluronic acid (HA) has been by BestBio Biotechnology Co., Ltd. (Shanghai, China). Lyso Tracker
widely used as a targeted “guider” in tumor cells with CD44 re- Red probe was acquired from G-clone Biotechnology Co., Ltd. (Bei-
ceptor overexpression [30,31], therefore, HA modification applied jing, China).
to ZIF-8 based nanoparticles can specifically increase cellular up-
take [32–35] and thus improve therapeutic efficacy. 2.2. Synthesis of CuS-ZIF-8 and CPT-CuS-ZIF-8
Microneedles (MNs) are a novel transdermal drug delivery sys-
tem (DDS), which can pierce the stratum corneum painlessly and CuS nanoparticles were prepared by the hydrothermal method.
generate mechanical microchannels to remarkably facilitate drug First, 112 mg of CuCl2 ·2H2 O was added into 80 mL of aqueous solu-
permeation in superficial tumor therapy. Dissolving microneedles tion of PVP-K30 (4 mg/mL) and stirred for 30 min, and then 0.8 mL
(DMNs) are the most extensively explored type of MNs during the of aqueous solution of Na2 S (60 mg/mL) was poured into the above
past years [36–38], because DMNs can encapsulate drug molecules solution. After 5 min, the sample was heated at 90 °C for 15 min
into the whole needle with high and precise drug dose, and re- to prepare CuS nanoparticles. Afterwards, 8 mL of as-prepared CuS
lease drug gradually after being dissolved in the tissue fluid. In nanoparticle methanol dispersion (1.63 mg/mL) was mixed with 20
addition, compared with intravenous injection, DMNs can realize mL of Zn(NO3 )2 ·6H2 O methanol solution (293 mg, 0.98 mmol). Af-
uniform three-dimensional channel distribution, improve drug en- ter stirring for 25 min, 12 mL of 2-Mim methanol solution (649
richment in tumors, and minimize systematic side effects [37–40]. mg, 7.90 mmol) was added into the aforementioned mixture, and
However, the direct loading of drug molecules into DMNs consists the mixture was sequentially stirred for another 25 min. CuS-ZIF-8
of many problems, such as incompatible properties between drugs nanoparticles were collected by centrifuging at 12,0 0 0 rpm for 5
and polymeric materials, uncontrollable drug release, and unsat- min. Subsequently, the nanoparticles were washed with methanol
isfactory efficacy of monotonic drug delivery [41]. Therefore, it is to remove redundant reagents for several times, and then were re-
urgent and necessary to develop a stable, intelligent, and multi- dispersed in 3 mL of methanol for storage.
modal DDS to overcome these drawbacks. The only difference of the synthesis of CPT-CuS-ZIF-8 was that
We are inspired to design a multifunctional nanoparticle- CPT (20 mg, 0.06 mmol) was pre-dissolved into the Zn(NO3 )2 ·6H2 O
integrated DMNs for multi-modal collaborative therapy of methanol solution. In the process, CuS nanoparticles were embed-
melanoma. This system can co-load various drugs, improve ded in ZIF-8 framework and CPT was in situ trapped into the pores
the specific uptake and distribution of tumor, and achieve intel- of the hybrid framework during crystal growth [43,44], thereby
ligent and spatiotemporal drug release, thereby promising a high forming CPT-CuS-ZIF-8 in one step. The supernatant during the
efficiency with low-dose administration. First, we simultaneously synthesis of CPT-CuS-ZIF-8 was collected for the measurement of
encapsulated the photothermal agent (CuS nanoparticles) and hy- encapsulation efficiency (EE) and drug loading capacity (DLC) via

165
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Scheme 1. Schematic illustration of (A) preparation procedure of CPT-CuS-ZIF-8@HA and (B) multifunctional nanoparticle-integrated DMNs for local administration, targeted
delivery, intelligent drug release, and synergetic chemo-photothermal therapy in melanoma treatment.

high performance liquid chromatograph (HPLC). EE and DLC were nitrogen adsorption-desorption analysis (ASAP2460, Micromeritics,
calculated according to the following equations: America). Crystallography was analyzed by PXRD (D8 Advance,
  Bruker, Karlsruhe, Germany) using Cu Kα radiation with 2θ in the
EE(% ) = Minitial drug − Mdrug in supernatant /Minitial drug × 100%
  range of 3o -40o at a scanning rate of 5o /min. Characteristic of mass
DLC(% ) = Minitial drug − Mdrug in supernatant /Mnanoparticles × 100% varying with temperature was monitored by TGA (TG209F1 libra,
NETZSCH-Gerätebau GmbH, Germany).
2.3. Preparation of CPT-CuS-ZIF-8@HA
2.5. In vitro photothermal effect of nanoparticles
CPT-CuS-ZIF-8@HA was prepared by HA surface modification on
CPT-CuS-ZIF-8 via coordination bond [45] and electrostatic interac-
One milliliter of CuS, CuS-ZIF-8, and CPT-CuS-ZIF-8 samples
tion [46]. First, 4.50 mg of HA was dissolved in 6 mL of water,
with the same CuS concentration (50 μg/mL), and 1 mL of CPT-
and 1.5 mL of as-prepared CPT-CuS-ZIF-8 nanoparticles (32 mg)
CuS-ZIF-8@HA samples with different concentrations (10 0, 30 0,
methanol dispersion was diluted with water to 3 mL. Afterwards,
and 500 μg/mL) were put in 2 mL Eppendorf tube, and subse-
the aforementioned HA water solution (0.50 mg/mL) and the CPT-
quently irradiated by an 808 nm laser (1.0 W/cm2 ) for 5 min. The
CuS-ZIF-8 nanoparticles methanol/water dispersion (3.56 mg/mL)
temperature was measured and photothermal images were taken
were mixed and stirred for 30 min. Subsequently, CPT-CuS-ZIF-
by an infrared thermal sensing and imaging equipment (TiS75,
8@HA nanoparticles were collected by centrifuging at 15,0 0 0 rpm
Fluke, Everett, WA, America) every 30 s. ZIF-8 nanoparticles were
for 3 min. Finally, the nanoparticles were washed with water for
used as a contrast.
several times, and then were redispersed in 1 mL of water for stor-
To evaluate the photostability, the CPT-CuS-ZIF-8@HA and pris-
age.
tine CuS suspension were exposed to NIR for three cycles. Each cy-
The fluorescein labeled nanoparticles (FL-CuS-ZIF-8 and FL-CuS-
cle was composed of 5 min irradiation followed by nearly 20 min
ZIF-8@HA) were also synthesized for cellular uptake, subcellular
for cooling down.
location, and in vivo imaging and biodistribution studies using the
similar procedure mentioned above.
2.6. In vitro drug release
2.4. Characterization
To investigate the low pH and laser-responsive drug release of
Particle size and zeta potential were measured by DLS (Zeta- CPT-CuS-ZIF-8@HA, the nanoparticles were dispersed in 6 mL of
sizer Nano, Malvern Instrument, Britain). Morphology was char- PBS solution with various pH values (pH 5.0, 6.0 and 7.4), and then
acterized by transmission electron microscopy (TEM, FEI Tecnai the samples were incubated with continuous shaking (100 rpm) at
G2 Spirit, Netherlands) and Field-Emission-SEM (GeminiSEM500, 37 °C. At predetermined time points, 1.0 mL of supernatant was
Zeiss/Bruker, Germany). The molecular interaction was indicated collected and equal volume of corresponding fresh PBS solution
by FT-IR (UATR Two, Perkin Elmer, America). The UV-Vis spec- was added. Meanwhile, the samples were irradiated with an 808
trum was measured by a UV-vis spectrophotometer (UV-2600, Shi- nm laser (1.0 W/cm2 ) for 5 min, followed by similar sampling pro-
madzu, Japan). Surface area and pore volume were evaluated by cess. Finally, the CPT concentration was measured by HPLC.

166
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

2.7. Cell culture h. Finally, the cells were collected and handled according to the in-
struction of the kit, and subsequently were quantitatively analyzed
B16 cells (a cell line of mouse melanoma cells) were cul- via flow cytometry.
tured with the 1640 medium. A375 cells (a cell line of human
melanoma cells) were cultured with the DMEM medium. Both cell
2.10. Fabrication and characterization of CPT-CuS-ZIF-8@HA@MN
culture medium contained 10% FBS and 1% antibiotics (penicillin
and streptomycin). An incubator with 5% CO2 was used to culture
The nanoparticle-integrated microneedles were prepared by
cells at 37 °C.
multi-step centrifugation casting method referring to a previous
report [47]. First, the tip solution was made of PVP K30 and PVA
2.8. In vitro cellular uptake and subcellular location
103 with a ratio of 6:1 (w/w), and the base solution was 30% (w/v)
PVP K90 prepared by pre-swelling in ethanol overnight. And then,
B16 cells were pre-cultured in NUNCTM imaging 24-well plate
a quarter volume of tip solution was added to the CPT-CuS-ZIF-
for 24 h at 37 °C. The excessive amount of HA medium solution (10
8@HA nanoparticle suspension with a concentration of 16 μg/mL
mg/mL) was added into the plates of HA+FL-CuS-ZIF-8@HA group
to form tip mixture. The preparation procedure of CPT-CuS-ZIF-
for 2 h pre-incubation. After that, fresh FL-CuS-ZIF-8 and FL-CuS-
8@HA@MN was described below. Proper volume of tip mixture was
ZIF-8@HA medium suspension were added to replace the original
spread out on the poly (dimethylsiloxane) (PDMS) female mold and
medium in plates for 4 h incubation. Then, the cells were rinsed
centrifuged at 40 0 0 rpm for 5 min at 4 °C. Then, excess tip mix-
three times with cold PBS solution and fixed by 4% paraformalde-
ture on the surface of female mold was removed and the mold was
hyde at 4 °C for 10 min in dark. Later, the fixed liquid was re-
centrifuged at 40 0 0 rpm for 1 h to enrich the nanoparticles to tip.
moved, and the cells were rinsed with PBS again and stained by
Subsequently, the female mold was dried for 10 h in a dryer at
DAPI for 15 min. Finally, the cells were rinsed and fluorescence
room temperature, and the operation of centrifugation and drying
imaging was performed by FV30 0 0 Olympus confocal microscope
was repeated to improve drug loading. Afterwards, the tip solution
for cellular uptake observation. As for flow cytometry analysis, the
and base solution were poured on the female mold and then cen-
cells in plates were washed and collected to obtain quantitative re-
trifuged for 5 min in sequence. Finally, the microneedle patch was
sults of cellular internalization. Additionally, the lysosome imaging
collected after drying for 24 h in a dryer at room temperature. The
was performed with the aid of LysoTracker Red probe for subcel-
CPT loading amount of CPT-CuS-ZIF-8@HA@MN was calculated by
lular location study. After 2 h of incubation with nanoparticle sus-
HPLC after dissolving the whole excisional microneedle tips.
pension, the cells were rinsed three times with PBS solution, and
In addition, the morphology of DMNs was characterized un-
treated with pre-heated LysoTracker Red probe to stain lysosome
der a mobile microscope and Thermal Field Emission Enviromen-
at 37 °C for 30 min in dark. Later, the red probe solution was re-
tal SEM EDS EBSD (Quanta 400F, FEI/OXFORD/HKL, Netherlands).
moved and the cells were rinsed with PBS and stained by Hoechst
The nanoparticle distribution and 3D reconstruction were observed
for 15 min.
through FV30 0 0 Olympus confocal microscope with FL-CuS-ZIF-
8@HA@MN.
2.9. In vitro cytotoxicity, live/dead cell staining, and apoptosis assay

In vitro anti-tumor efficiency of CPT-CuS-ZIF-8@HA was eval- 2.11. Insertion capability evaluation
uated by a standard CCK-8 method. B16 cells (12,0 0 0 cells/well)
and A375 cells (8,0 0 0 cells/well) were seeded in 96-well plate and To evaluate the insertion capability, the CPT-CuS-ZIF-8@HA@MN
cultured for 24 h. After 4 h of incubation with nanoparticle sus- was pressed on the isolated depilated skin of rat and porket for 2
pension of different concentrations and following 5 min NIR irra- min. After removal, the insertion pores were imaged with a mo-
diation (1.0 W/cm2 ), the nanoparticle medium suspension was re- bile microscope. Furthermore, the inserted skin was also stained
moved and the cells were cultured in fresh medium for another 20 by hematoxylin and eosin (H&E).
h. Subsequently, the cells were incubated with 110 μL/well of CCK-
8 solution (CCK-8: medium = 1:10) for 1 h at 37 °C. Finally, the
2.12. In vitro photothermal effect of DMNs
absorbance of each well at 450 nm was measured by a microplate
reader. The cell viability was calculated according to the following
Blank DMN, CuS-ZIF-8@MN, and CPT-CuS-ZIF-8@HA@MN were
Equation:
  irradiated by an 808 nm laser with different power intensities (1.0,
Cell Viability (% ) = Asample − Ablank /(Acon − Ablank ) × 100% 0.5, 0.3 W/cm2 ) for 5 min. The temperature was measured and
photothermal images were taken every 30 s. The blank DMN was
The live/dead cell staining assay was studied via a Cell Imager
applied as a contrast.
(EVOS FL Auto, Life Technologies, America) after being stained by
To investigate the photostability of nanoparticle-integrated
the Calcein-AM/PI kit. B16 cells (10 0,0 0 0 cells/well) were seeded
DMNs, the CPT-CuS-ZIF-8@HA@MN was exposed to NIR (1.0, 0.5
in 24-cell plate and pre-cultured for 24 h. After 4 h of incubation
W/cm2 ) for three cycles. Each cycle was composed of 5 min irradi-
with nanoparticle suspension (CPT: 11.38 μg/mL, CuS: 9.85 μg/mL)
ation followed by 7 min or 4 min for cooling down.
and following 5 min NIR irradiation (1.0 W/cm2 ), the nanoparticle
medium suspension was removed and the cells were rinsed with
fresh medium twice. Finally, cells in each well were stained accord- 2.13. Construction of animal melanoma models
ing to the instruction of the kit, and subsequently observed via a
Cell Imager. C57BL/6 female mice were supplied by Guangdong Medical Ex-
The cell apoptosis assay was investigated using flow cytometry perimental Animal Center, and all experiments were supervised by
after being stained by the Annexin V-FITC/PI apoptosis assay kit. the Animal Ethics Committee of Sun Yat-sen University (Approval
B16 cells (20 0,0 0 0 cells/well) were seeded in 6-well plate and pre- No. SYSU-IACUC-2021-0 0 0 029, Guangdong, China). After adaptive
cultured for 24 h. After 4 h of incubation with nanoparticle sus- feeding for 7 days in the Experimental Animal Center of Sun Yat-
pension (CPT: 3.41 μg/mL, CuS: 2.96 μg/mL) and following 10 min sen University (Guangdong, China), the suspension of B16 cells (50
NIR irradiation (1.0 W/cm2 ), the nanoparticle suspension was re- μL, 3 × 106 ) was inoculated subcutaneously into the depilated back
moved and the cells were cultured in fresh medium for another 20 of the mice.

167
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

2.14. In vivo photothermal effect the tissues were sliced for different tests. The tumor slices were
stained with H&E, cleaved caspase 3, terminal deoxynucleotidyl
The tumor-bearing C57BL/6 female mice were divided into transferase (TdT)-mediated dUTP nick end labeling (TUNEL), and
four groups randomly after the tumor volume being 100-120 Ki67 to assess the antitumor efficacy, and the major organs slices
mm3 : (1) control+NIR, (2) CuS-ZIF-8@MN+NIR, (3) CPT-CuS-ZIF- were stained by H&E for safety evaluation.
8@HA@MN+NIR, and (4) CPT-CuS-ZIF-8@HA (I.T.)+NIR. The corre-
sponding DMN patches were pressed on the tumor site for 2 min
2.19. Statistical analysis
by thumb and subsequently secured on the back with medical
tape. Simultaneously equal amount of CPT-CuS-ZIF-8@HA nanopar-
Data are expressed as the means ± standard deviation (SD).
ticles were intratumorally injected. After 4 h, the patches were
Statistical analysis was performed using a one-way ANOVA test
teared and the tumor site was irradiated by an 808 nm laser (1.0
(Graphpad Prism 7.00). The post hoc comparisons of the means of
W/cm2 ) for 5 min. The temperature was monitored and photother-
individual groups were performed using least significant difference
mal images were taken every 1 min.
test. Differences were considered significant if P < 0.05.
2.15. In vivo imaging and biodistribution
3. Results and discussion
The administration of FL-CuS-ZIF-8@HA@MN was almost simi-
lar to the aforementioned in vivo photothermal study. Live fluores- 3.1. Preparation and characterization of CPT-CuS-ZIF-8@HA
cence imaging system (IVIS Lumina XRMS, PerkinElmer, America)
was used to detect the fluorescence signals at 0, 2, 4, 6, 8, and The preparation procedure of CPT-CuS-ZIF-8@HA was illustrated
12 h. Finally, the mice were sacrificed after the last time point, in Scheme 1A. First, one of the guests, CuS nanoparticles, were syn-
and the fluorescence signal distribution in tumor and major organs thesized with an extremely small size about 10 nm (Fig. S1) via
(heart, liver, spleen, lung, and kidney) was observed. hydrothermal method. Then, CuS and CPT were co-encapsulated in
Additionally, the drug penetration capability was investigated. ZIF-8 and followed by HA coating to prepare CPT-CuS-ZIF-8@HA.
After administration of FL-CuS-ZIF-8@HA@MNs for 4 h, the mice The elemental mapping confirmed CuS was distributed within the
were sacrificed and the excised tumor were quickly frozen in liquid framework of ZIF-8 (Fig. S2). As observed by SEM (Fig. 1A), the
nitrogen. Afterwards, the frozen sections of tumor were stained by morphology of CPT-CuS-ZIF-8@HA was roughly spherical, and their
DAPI and subsequently imaged by FV30 0 0 Olympus confocal mi- diameters were approximately 50 nm, which was consistent with
croscope. the average hydrodynamic diameter determined by DLS (Fig. 1B).
For comparison, the morphology and hydrodynamic diameter of
2.16. In vivo anti-tumor study
ZIF-8, CuS-ZIF-8, and CPT-CuS-ZIF-8 were tested and exhibited in
Fig. S1 and Fig. S3A, respectively. The stepwise increment of par-
The tumor-bearing C57BL/6 female mice were divided into six
ticle size was attributed to the guest encapsulation and surface
groups randomly after the tumor volume being 100-120 mm3 :
modification during different synthetic stages. In addition, the zeta
(1) untreated, (2) CuS-ZIF-8@MN, (3) CuS-ZIF-8@MN+NIR, (4) CPT-
potential was measured and exhibited in Fig. S3B. The potential of
CuS-ZIF-8@HA@MN, (5) CPT-CuS-ZIF-8@HA nanoparticles intratu-
ZIF-8 was +18.8 eV and slightly decreased to +16.4 eV for CuS-ZIF-
moral injection (I.T.)+NIR, and (6) CPT-CuS-ZIF-8@HA@MN+NIR.
8. After CuS and CPT co-encapsulation, a significant change was
The administration of DMNs was according to the aforementioned
observed from +18.8 eV (ZIF-8) to +6.0 eV (CPT-CuS-ZIF-8). Fi-
in vivo photothermal study. Meanwhile, equal amount of CPT-CuS-
nally, HA modification dramatically reversed the zeta potential to
ZIF-8@HA nanoparticles were intratumorally injected. After 4 h, the
-20.6 eV (CPT-CuS-ZIF-8@HA). This zeta potential variation initially
tumor site was irradiated by an 808 nm laser (1.0 W/cm2 ) for 5
demonstrated the successful incorporation of negatively charged
min for NIR groups. Subsequently, tumor volume and body weight
CuS nanoparticles and CPT and the successful coating of negatively
were measured and recorded every two days. Finally, the mice
charged HA. The negatively charged surface also endows CPT-CuS-
were sacrificed, and the tumor and major organs were weighed af-
ZIF-8@HA with favorable stability for further biological tests.
ter excising on day 12. During the experiment, the mice would be
Then, FTIR spectrum further qualitatively ascertained the mod-
sacrificed for kindness when tumor volume was over 20 0 0 mm3 .
ification processes of the nanoplatform. As presented in Fig. 1C,
The tumor volume was calculated according to the following equa-
the vibrations between CuS-ZIF-8 and ZIF-8 were nearly identical
tion:
except for the weak peak at 1701 cm−1 from the C=O stretch-
Tumor volume = L(tumor length ) × W 2 (tumor width ) /2 ing vibration of PVP existed in CuS nanoparticles, which implied
In addition, the survival rates of the mice were monitored every the existence of CuS nanoparticles. The stretching bands vibra-
day in four weeks. tions in CPT-CuS-ZIF-8 corresponding to C-N (1097 cm−1 ) and C-
O (1235 cm−1 ) were indicative of CPT. In addition, the new broad
2.17. Safety evaluation peak around 3310 cm−1 of CPT-CuS-ZIF-8@HA could be ascribed
to the stretching vibration of O-H of HA. The above representa-
To evaluate the safety of DMNs administration, healthy tive signals further demonstrated the successful preparation of the
C57BL/6 female mice were divided into three groups randomly: nanoparticles.
(1) untreated, (2) CPT-CuS-ZIF-8@HA@MN, and (3) CPT-CuS-ZIF- The UV-Vis spectra of various samples were displayed in
8@HA@MN+NIR. After DMNs administration and following NIR ir- Fig. 1D. CPT and ZIF-8 did not present obvious absorption between
radiation (1.0 W/cm2 ) for 5 min, the skin condition was observed 600 and 1000 nm, while samples containing CuS exhibited a broad
and imaged by a digital camera at predetermined date (day 0, 1, 3, absorption band in this scope. In addition, compared with other
5, 7, and 10). nanoparticles, CPT-CuS-ZIF-8 and CPT-CuS-ZIF-8@HA displayed an
obvious absorption peak of CPT at around 367 nm. These results
2.18. Histology and pathological investigation proved the successful encapsulation of guests. Simultaneously, the
concentrations of CPT were determined by an UV-detector in HPLC
The excised tumors and major organs were fixed in 4% for calculating EE and DLC. And the EE and DLC were determined
paraformaldehyde for 24 h before paraffin embedding, and then to be 74.18% and 11.38%, respectively.

168
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 1. (A) SEM image and (B) size distribution of CPT-CuS-ZIF-8@HA. (C) FT-IR spectra of CPT-CuS-ZIF-8@HA, CPT-CuS-ZIF-8, CuS-ZIF-8, HA, CPT, CuS, and ZIF-8. (D) UV
absorption spectra of CPT-CuS-ZIF-8@HA, CPT-CuS-ZIF-8, CuS-ZIF-8, CPT, CuS, and ZIF-8. (E) Nitrogen adsorption-desorption isotherms and (F) pore size distribution of ZIF-8,
CuS-ZIF-8, and CPT-CuS-ZIF-8. (G) PXRD patterns and (H) thermogravimetric analysis of CPT-CuS-ZIF-8@HA, CPT-CuS-ZIF-8, CuS-ZIF-8, HA, CPT, CuS, and ZIF-8.

Nitrogen adsorption-desorption analysis was utilized to calcu- 3.2. In vitro photothermal effect and drug release properties of
late the surface area and pore volume of different samples. As ex- CPT-CuS-ZIF-8@HA
hibited in Fig. 1E, the BET surface area of ZIF-8, CuS-ZIF-8, and
CPT-CuS-ZIF-8 were 1886, 1321, 1289 m²/g, respectively. The sur- Different samples with equal CuS concentration (50 μg/mL)
face area decreased with the encapsulation of CuS nanoparticles were irradiated to study the photothermal effect. After irradiation,
and CPT, because the non-porous CPT and CuS nanoparticles con- similar to the pure CuS, the temperature of CPT-CuS-ZIF-8@HA in-
tributed to the mass of the composite and simultaneously ob- creased from 28.2 °C to 57.4 °C, while the temperature increment
structed the nitrogen flow [43]. Pore size distribution of the three of ZIF-8 was negligible (Fig. 2A and B). The above results indicated
samples all exhibit a maximum at 1 nm (Fig. 1F), which indicated that the preparation procedure did not affect the photothermal
the encapsulation could arise negligible influence on the pore conversion efficacy of CuS, and the wonderful temperature-rising
size. performance of CPT-CuS-ZIF-8@HA made it potential to apply for
PXRD can identify the crystallography of samples. As shown in further PTT. Moreover, CPT-CuS-ZIF-8@HA displayed concentration-
Fig. 1G, the diffraction peaks of CPT-CuS-ZIF-8 and CPT-CuS-ZIF- dependent photothermal effects (Figs. 2C and S4). Therefore, the
8@HA were consistent with the characteristic peaks of ZIF-8, sug- intensity of PTT could be regulated on demand by dosing of CPT-
gesting that the incorporation of guests and HA modification did CuS-ZIF-8@HA. In addition, as shown in Fig. 2D, no obvious change
not affect the crystal structure. However, the characteristic diffrac- was observed in the temperature profiles among the three on/off
tion peaks related to CPT disappeared in the PXRD patterns of NIR irradiation cycles, which suggested that the nanoparticles pos-
CPT-CuS-ZIF-8, which indicated that CPT, as a guest molecule, was sessed superior photothermal stability for repeated and pulsatile
loaded into ZIF-8 pores in amorphous or molecular state. Further- therapy [42].
more, the successful preparation process was further verified by In addition, the drug release properties were investigated.
the weight loss observed in TGA (Fig. 1H). As photothermal agent, CuS nanoparticles were embedded in

169
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 2. (A) Temperature change curves of CPT-CuS-ZIF-8@HA, CPT-CuS-ZIF-8, CuS-ZIF-8, CuS, and ZIF-8 under laser irradiation (1.0 W/cm2 ) at different time points (equal CuS
concentration of 50 μg/mL). (B) The thermographic images of ZIF-8 and CPT-CuS-ZIF-8@HA (500 μg/mL) under NIR irradiation (1.0 W/cm2 ) at different time points (equal CuS
concentration of 50 μg/mL). (C) Temperature change curves of CPT-CuS-ZIF-8@HA at different concentrations under NIR irradiation (1.0 W/cm2 ). (D) Photothermal conversion
stability curves of CuS and CPT-CuS-ZIF-8@HA for three cycles (1.0 W/cm2 , equal CuS concentration of 50 μg/mL). (E) CPT release profiles in PBS with different pH values
under on/off NIR irradiation (1.0 W/cm2 ) cycles (mean±S.D., n=3).

the framework of ZIF-8. When CPT-CuS-ZIF-8@HA was irradi- FL-CuS-ZIF-8@HA) via fluorescence imaging and flow cytometry
ated by NIR, the internal CuS could convert light energy to heat analysis. The fluorescence images (Fig. 3A) under FITC channel and
[11,35,48,49]. However, the other components were not heated si- quantitative analysis (Fig. 3B) indicated that the fluorescence in-
multaneously for their low light absorption coefficient. The un- tensity from B16 cells incubated with FL-CuS-ZIF-8@HA was sig-
even heating resulted in uneven volume change in different re- nificantly higher than that of FL-CuS-ZIF-8 group, and which de-
gions on account of thermal expansion in the whole CPT-CuS-ZIF- creased obviously when B16 cells were preincubated with an ex-
8@HA, which could cause the structure collapse, thereby facilitat- cessive amount of HA [51], confirming that HA coating notably
ing the release of CPT. In Fig. S5, the results of TEM confirmed enhanced the cellular uptake of the nano-DDS. The quantitative
that laser irradiation could cause the structure collapse of CPT- results of flow cytometry analysis (Fig. 3C) reconfirmed the en-
CuS-ZIF-8@HA. Moreover, since the coordination bonds between hanced cellular uptake through HA. To further investigate the cel-
zinc ions and 2-Mim linkers would be broken for protonation of lular uptake route of FL-CuS-ZIF-8@HA, B16 cells were stained
2-Mim in acidic condition [50], the drug release of CPT-CuS-ZIF- with two fluorescent tracers, LysoTracker Red probe and Hoechst,
8@HA was speculated to be pH-responsive. To evaluate the drug to trace the intracellular lysosome and nucleic acid (nuclear), re-
release behaviors, CPT-CuS-ZIF-8@HA was irradiated repeatedly by spectively (Figs. 3D and S6). The green fluorescence of FL-CuS-ZIF-
an 808 nm laser for 5 min at predetermined time points in release 8@HA matched well with the red fluorescence from lysosome af-
medium (PBS) with different pH. The PBS with the pH of 5.0, 6.0, ter incubation for 2 h (Fig. 3D), and the intensity enhanced grad-
7.4 were designed to simulate lysosomes, tumorous microenviron- ually with the extension of incubation time (Fig. S6). These re-
ment, and normal tissue fluid, respectively. As presented in Fig. 2E, sults demonstrated that FL-CuS-ZIF-8@HA was internalized to tu-
a burst release was appeared with the laser irradiation, while the mor cell through lysosome-mediated endocytosis. Since the pH of
release without laser was really gradual, confirming the laser ac- intracellular lysosomes ranges from 4.5 to 5.0 [28,50], we specu-
celerated release effect. Furthermore, the release rate of CPT was lated that the pH-responsive drug delivery system would disinte-
significantly enhanced in acid medium due to the disintegration of grate well after internalization, thereby releasing the encapsulated
ZIF-8 [25]. The cumulative drug release amount in PBS with the drug molecules intelligently as designed.
pH of 5.0 was about 65% in 12 h, which was five times as much as
that in pH 7.4. Taken together, the release profiles confirmed the 3.4. In vitro cytotoxicity, live/dead cell staining, and apoptosis assay
pH-responsive and laser accelerated release properties of the DDS.
It could be predicted that the DDS would be further applied to var- The cytotoxicity was studied on B16 and A375 cells by CCK-8
ious tumor therapy, because acid tumor microenvironment and ad- assay. As displayed in Fig. 4A, the carrier (CuS-ZIF-8) exhibited no
justably external laser irradiation would control the drug release obvious toxicity at concentrations ranging from 6.25 to 100 μg/mL
intelligently and spatiotemporally at the lesion site on demand. on B16 cells, indicating its superior biocompatibility. In contrast,
the groups of CPT-CuS-ZIF-8@HA and CPT-CuS-ZIF-8@HA+NIR dis-
3.3. In vitro cellular uptake and subcellular location played high toxicity in a dose-dependent manner. For instance,
with the concentration of CPT-CuS-ZIF-8@HA increasing from 6.25
The cellular uptake and intracellular distribution were evalu- μg/mL (CPT: 0.72 μg/mL) to 100 μg/mL (CPT: 11.38 μg/mL), the
ated by fluorescein encapsulated nanoparticles (FL-CuS-ZIF-8 and cellular viability remarkably decreased from 68.8% to 23.7%, and

170
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 3. (A) Cellular uptake of FL-CuS-ZIF-8, FL-CuS-ZIF-8@HA, and HA + FL-CuS-ZIF-8@HA by B16 cells after incubation for 4 h. (B) Corresponding quantitative green fluo-
rescence intensities of B16 cells after incubation with FL-CuS-ZIF-8, FL-CuS-ZIF-8@HA, and HA + FL-CuS-ZIF-8@HA for 4 h (∗ ∗ P < 0.01 vs. FL-CuS-ZIF-8@HA, ∗ ∗ ∗ P< 0.001 vs.
FL-CuS-ZIF-8@HA, n=3). (C) Flow cytometry analysis of B16 cells incubated with FL-CuS-ZIF-8, FL-CuS-ZIF-8@HA, and HA + FL-CuS-ZIF-8@HA for 4 h, respectively. (D) The
fluorescence images of the location of FL-CuS-ZIF-8@HA in B16 cells after incubation for 2 h. The cell nuclei and lysosomes were labeled by Hoechst and LysoTracker Red,
respectively. Scale bars for (A) and (D): 50 μm.

it was much lower after NIR irradiation. Unexpectedly, the group age of CPT-CuS-ZIF-8@HA+NIR group, which further manifested
of CuS-ZIF-8+NIR showed low toxicity even at the concentration the synergistic therapeutic effect consistent with the aforesaid cy-
of 100 μg/mL. Previous studies verified that coagulative necrosis totoxicity and live/dead cells staining assay.
would occur when the temperature was above 50°C through cell
membrane collapse, protein denaturation, and mitochondrial dys- 3.5. Fabrication and characterization of CPT-CuS-ZIF-8@HA@MN
function [52–54]. While the heat-induced cellular injury under 45
°C could be reversed by tumor thermo-resistance at this concen- Systemic administration remains the most conventional mode
tration. In addition, the results of cytotoxicity on A375 cells were for tumor therapy, while the unspecific toxicity is still one of the
almost consistent with those on B16 cells (Fig. 4B). Taking all the greatest problems [41]. Therefore, topical drug delivery was intro-
above results together, the CPT-CuS-ZIF-8@HA+NIR group demon- duced for regional and superficial tumor therapy. DMNs are a kind
strated the highest cytotoxicity among all groups in both cell lines, of promising transdermal drug delivery (TDD) device in mediat-
which verified the nano-platform could achieve a synergistic effect ing a local delivery. In this research, the nanoparticle-integrated
for cancer therapy in vitro as predicted. Meanwhile, the results of microneedles were prepared by multi-step centrifugation casting
live/dead cell staining assay (Fig. 4C) were almost in concert with method (Fig. S7). The patch was composed of 12 × 12(144) mi-
the synergistic cell inhibition effect of cytotoxicity study. croneedles with 1200 μm in height, and the CPT loading amount
Besides, the apoptosis assay was performed to evaluate the was 74 μg per patch. The morphology of DMNs was observed by
cell apoptosis. As exhibited in Fig. 4D, Q2 represents late apopto- SEM, optical microscope, and confocal laser scanning microscopy
sis or necrosis cells, and Q4 represents viable cells. The apoptotic (CLSM). As shown in Fig. 5A, the tips of both blank DMNs and
ratios of cells treated by CuS-ZIF-8+NIR and CPT-CuS-ZIF-8@HA nanoparticle-integrated DMNs exhibited as the rectangular pyra-
were 8.99% and 18.9%, respectively, which were significantly lower mid, indicating that the encapsulation of nanoparticles did not
than that of CPT-CuS-ZIF-8@HA+NIR group (40.1%). The quantita- cause significant morphology change or deficiency on tips. In ad-
tive analysis demonstrated the highest cellular apoptotic percent- dition, the optical image (Fig. 5B), fluorescent image (Fig. 5C), and

171
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 4. Cell viability of (A) B16 and (B) A375 cells after incubation with various samples for 24 h (mean±S.D., n=6). (C) Fluorescence images of B16 cells with various
treatments co-stained with Calcein-AM/PI. Scale bar: 200 μm. (D) Flow cytometric analysis of apoptosis on B16 cells after incubation with various samples.

3D reconstruction (Fig. 5D) further confirmed the homogenous dis- DMNs was a vital factor for effective TDD, which could be eval-
tribution of nanoparticles in the tips of FL-CuS-ZIF-8@HA@MN. The uated by isolated depilated skin of rat and porket. After puncture
tips enrichment of nanoparticles made it prospective to realize the of DMNs, the microchannels left in rat skin (Fig. 5E) and porcine
efficient and precise drug delivery after DMNs puncture and thus skin (Fig. S8A) were clearly observed via an optical microscope.
enhance efficacy. Apart from that, the skin insertion capacity of Additionally, H&E staining of the inserted skin (Figs. 5F and S8B)

172
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 5. (A) SEM images of blank DMN, CuS-ZIF-8@MN, and CPT-CuS-ZIF-8@HA@MN. Scale bar: 500 μm. (B) Photograph of CPT-CuS-ZIF-8@HA@MN. (C) CLSM images of
FL-CuS-ZIF-8@HA@MN. Scale bar: 300 μm. (D) 3D reconstruction image of FL-CuS-ZIF-8@HA@MN under CLSM. (E) Photograph of the excised rat skin after insertion of
CPT-CuS-ZIF-8@HA@MN. (F) H&E staining histological section of inserted rat skin. Scale bar: 200 μm.

further verified that the nanoparticle-integrated DMNs were strong matically elevate to around 50 °C in only 1 min irradiation fol-
enough to penetrate cutaneous barrier for drug delivery. lowed by gradual elevation (Fig. 7A and B). Previous studies have
To investigate the in vitro photothermal effect of CPT-CuS- proved that temperatures above 48 °C could induce a drastic and
ZIF-8@HA@MN, the DMNs were irradiated by an 808 nm laser nonreversible activation of cell apoptosis pathway to kill tumor
with different power intensities (Figs. 6A, B, S9A, and S9B). cells [17]. Therefore, CPT-CuS-ZIF-8@HA@MN was expected to be
As displayed in Fig. 6A and B, compared with blank DMNs, an effective system for further in vivo PTT on account of its supe-
the temperature of CPT-CuS-ZIF-8@HA@MN rapidly elevated to rior in vivo photothermal ability.
∼60 °C from room temperature only in 1 min irradiation (0.5 In order to investigate the duration of FL-CuS-ZIF-8@HA at tu-
W/cm2 ) and subsequently remained steady, which demonstrated mor site, the fluorescence signals of mice were detected at differ-
the nanoparticle-integrated DMNs possessed satisfactory pho- ent time points after administration of FL-CuS-ZIF-8@HA@MN. As
tothermal effect. Fig. 6C indicated their power intensity-dependent shown in Fig. 7C, the fluorescence signals were mainly concen-
photothermal property. Moreover, in photothermal stability anal- trated at the tumor site. Quantitative analysis demonstrated that
ysis (Figs. 6D, S9C), the temperature variation curve of three laser the fluorescence intensity at the tumor site kept obvious during
on/off cycles were nearly identical. The excellent photothermal sta- 12 h, indicating the formulation had a long duration at adminis-
bility of CPT-CuS-ZIF-8@HA@MN endowed the DDS with the char- tration site (Fig. 7D). The long duration might be attributed to the
acteristics of repeated PTT and spatiotemporally controlled pul- formative drug reservoirs for continuous drug release in tumor af-
satile drug release for further application. ter DMNs administration. To further investigate the biodistribution
of FL-CuS-ZIF-8@HA, the mice were sacrificed at 12 h, and the flu-
3.6. In vivo photothermal effect and biodistribution orescence signal distribution in the excised tumor and major or-
gans (heart, liver, spleen, lung, and kidney) was observed. Figs. 7E
In vivo photothermal effect was performed on B16 tumor- and S10 showed that the fluorescence signal was nearly enriched
bearing mice at 4 h post-administration. Compared with control at the tumor site without appearing in major organs, suggesting
group, the localized temperatures in other three groups could dra- that the nanoparticles were difficult to exert adverse system effects

173
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 6. (A) The thermographic images of blank DMN, CuS-ZIF-8@MN, and CPT-CuS-ZIF-8@HA@MN under NIR irradiation (0.5 W/cm2 ) at different time points. (B) Temperature
change curves of blank DMN, CuS-ZIF-8@MN, and CPT-CuS-ZIF-8@HA @MN under NIR irradiation (0.5 W/cm2 ). (C) Temperature change curves of CPT-CuS-ZIF-8@HA@MN
under laser irradiation with different power intensities. (D) Temperature change curve of CPT-CuS-ZIF-8@HA@MN during three laser on/off cycles (0.5 W/cm2 ).

but dedicated to function at lesion site after DMNs administration. groups, indicating that the combination of chemotherapy and PTT
In addition, after FL-CuS-ZIF-8@HA@MN administration, the corre- realized a synergistic effect on melanoma inhibition in vivo. The
sponding tumor was excised for frozen sections to inspect the drug satisfactory synergistic effect might be attributed to the follow-
penetration capability. Fig. 7F displayed that the drug (green fluo- ing two reasons: First, the hyperthermia of PTT directly induced
rescence) could be delivered to deep tumor effectively. Taken to- thermal ablation of tumor cells; Second, ZIF-8 decomposition un-
gether, these results suggested the topical DMNs delivery was a fa- der acid tumor microenvironment and external NIR irradiation in-
vorable and promising route for local tumor therapy. duced an intelligent and spatiotemporal drug release to improve
cell killing. Except that, the antitumor efficacy in different admin-
3.7. In vivo anti-tumor study istration routes was also investigated. Compared with DMNs group,
the tumors of intratumoral injection group recurred on day 4 and
Inspired by the in vitro pharmacodynamics, the in vivo anti- grew larger, revealing the superiority of DMNs administration. This
tumor study was carried out, and the experimental process was might because the nanoparticles with liquid formulation could eas-
illustrated in Fig. 8A. The tumor-bearing C57BL/6 female mice ily diffuse into the surrounding tissues, thereby resulting in re-
were randomly divided into six groups (n=6) after the tumor duced drug accumulation at the tumor site after mono-point injec-
volume being 100-120 mm3 : (1) untreated, (2) CuS-ZIF-8@MN, tion. While after CPT-CuS-ZIF-8@HA@MN administration, hundreds
(3) CuS-ZIF-8@MN+NIR, (4) CPT-CuS-ZIF-8@HA@MN, (5) CPT-CuS- of microchannels would deliver nanoparticles into tumor with a
ZIF-8@HA nanoparticles intratumoral injection (I.T.)+NIR, and (6) three-dimensional uniform distribution, and the remained dissolv-
CPT-CuS-ZIF-8@HA@MN+NIR. The tumor volume curves (Fig. 8B) ing needles in skin could also act as drug reservoirs for continuous
showed that the volume of untreated group and CuS-ZIF-8@MN drug release.
group grew dramatically to nearly 20 0 0 mm3 on day 8 indicat- To further evaluate the anti-tumor effects, the tumors ex-
ing that the CuS-ZIF-8@MN was incapable to inhibit tumor growth. cised from each group were photographed (Fig. 8C) and weighed
The mice in these two groups were sacrificed for kindness on day (Fig. 8D) at the end of treatments. The tumor weight of CPT-CuS-
10. By contrast, the tumor growth was suppressed to some extent ZIF-8@HA@MN+NIR group was the lowest among all groups. In ad-
in CuS-ZIF-8@MN+NIR and CPT-CuS-ZIF-8@HA@MN groups. What dition, the survival of CPT-CuS-ZIF-8@HA@MN+NIR group was sig-
is more, the tumor volume of CPT-CuS-ZIF-8@HA@MN group was nificant prolonged (Fig. 8E), further confirming the noticeable tu-
always smaller than that of CuS-ZIF-8@MN+NIR group, which re- mor inhibiting ability of synergistic effect.
vealed the chemotherapy was slightly better than PTT in tumor in- Histological sections were performed to verify the anti-tumor
hibition. Such result was also consistent with the aforementioned effects at a microscopic level. H&E staining revealed the tumor tis-
in vitro cytotoxicity study. Moreover, the tumors in mice treated sues of untreated and CuS-ZIF-8@MN groups were compact and
with CPT-CuS-ZIF-8@HA@MN+NIR grew merely to 190 mm3 on integrated, while varying degrees of apoptotic signs, including nu-
the terminal day (day 12), which were always the smallest in six cleus pyknotic, cataclastic and dissolution of tumor cells, were ob-

174
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 7. (A) The thermographic images and (B) corresponding temperature change curves of control, CuS-ZIF-8@MN, CPT-CuS-ZIF-8@HA@MN, and CPT-CuS-ZIF-8@HA intratu-
moral injection (I.T.) groups under NIR irradiation (1 W/cm2 ) for different time points (mean±S.D., n=3). (C) Fluorescent images of B16 tumor-bearing mice after adminis-
tration of FL-CuS-ZIF-8@HA@MN. (D) Total fluorescent intensity of tumor site at different time intervals after administration of FL-CuS-ZIF-8@HA@MN (mean±S.D., n=3). (E)
Fluorescent image of major organs at 12 h post administration. (F) Fluorescence images of histological section of the tumor after administration with FL-CuS-ZIF-8@HA@MN.
Scale bar: 200 μm.

served in other groups (Fig. 8F). Furthermore, cell apoptosis state S11. The skin recovered to normal in only 1-3 days after dosing
of tumor tissues was also evaluated. The positive brown signal DMNs alone. While with a laser irradiation, the penetration site
in cleaved caspase 3 sections and green signal in TUNEL images was slightly red and scabbed on the first day. Fortunately, the scab
represented early apoptosis cells and late apoptosis cells, respec- faded away in 7-10 days. These results demonstrated the excel-
tively. Fig. 8F showed that the positive signals of apoptosis in CPT- lent safety of DMN administration and the reversible skin dam-
CuS-ZIF-8@HA@MN+NIR group were obviously higher than other age caused by irradiation. In addition, body weight variations were
groups, indicating the improved tumor apoptosis after synergistic monitored during the whole experimental process. No significant
treatment. Moreover, we examined the state of tumor cell pro- weight change was observed among the six groups in Fig. S12A,
liferation. Ki67 is a kind of proliferating cell associated antigen generally indicating the good biocompatibility and safety of the
that can be used as a marker to evaluate tumor cell proliferation formulation. Furthermore, the weight coefficient indexes and his-
[11,55,56]. The positive brown signal of Ki67 staining in CPT-CuS- tological analysis of major organs were also performed to eval-
ZIF-8@HA@MN+NIR group was the least in the six groups (Fig. 8F), uate the toxicity. The representative photos of major organs ex-
which demonstrated the superior inhibiting effect on tumor expan- cised from tumor-bearing mice were exhibited in Fig. S12B. The
sion. In general, histological assay confirmed that the great syner- weight coefficient indexes of major organs (heart, liver, lung, and
gistic efficacy in vivo was mainly attributed to promoting tumor kidney) among the six groups showed no noticeable difference ex-
cells apoptosis and inhibiting their proliferation. cept spleen (Fig. S12C). The spleen weight indexes in untreated and
Apart from the antitumor efficacy, we also assessed the safety CuS-ZIF-8@MN groups were higher than other groups, probably be-
of DMNs administration. The skin recovery photos of mice af- cause excessive tumor bearing may cause splenomegaly in mice.
ter CPT-CuS-ZIF-8@HA@MN administration were exhibited in Fig. Moreover, no obvious fibrosis, infiltration or inflammation was pre-

175
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

Fig. 8. (A) Schematic illustration of the experimental process. (B) Tumor volume changes of mice after various treatments for 12 days (mean±S.D., n=6). ∗ P < 0.05 vs.
CuS-ZIF-8@MN+NIR; ∗ ∗ P < 0.01 vs. CuS-ZIF-8@MN+NIR. (C) Photographs and (D) average weight of the excised tumors in different groups at Day 12. (mean±S.D., n=6).

P < 0.05 vs. CuS-ZIF-8@MN+NIR; ∗ ∗ P < 0.01 vs. CuS-ZIF-8@MN+NIR. (E) Survival rates of mice after various treatments for 28 days (n=6). ∗ P < 0.05 vs. Untreated;
∗∗
P < 0.01 vs. Untreated; ∗ ∗ ∗ P < 0.001 vs. Untreated. (F) Tumor section analysis by H&E, cleaved caspase 3, Ki67, and TUNEL staining. Scale bar: 400 μm.

176
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

sented in H&E staining of the major organs (Fig. S12D), reflecting [5] M.R. Ali, I.M. Ibrahim, H.R. Ali, S.A. Selim, M.A. El-Sayed, Treatment of natural
the excellent histological safety of the DDS. Based on the results mammary gland tumors in canines and felines using gold nanorods-assisted
plasmonic photothermal therapy to induce tumor apoptosis, Int. J. Nanomed.
of in vivo pharmacodynamics and safety, CPT-CuS-ZIF-8@HA@MN 11 (2016) 4849–4863.
demonstrated really high tumor inhibition rate and low toxicity, [6] H. Kim, K. Chung, S.J. Lee, D.H. Kim, H. Lee, Near-infrared light-respon-
which predicated our newly constructed DDS could be applied as sive nanomaterials for cancer theranostics, Wiley Interdiscip. Rev. Nanomed.
Nanobiotechnol. 8 (2016) 23–45.
an efficient platform for synergistic therapy against various super- [7] C. Yan, Q. Tian, S. Yang, Recent advances in the rational design of copper
ficial tumors. chalcogenide to enhance the photothermal conversion efficiency for the pho-
tothermal ablation of cancer cells, RSC Adv. 7 (2017) 37887–37897.
[8] L. Li, L.H. Rashidi, M. Yao, L. Ma, L. Chen, J. Zhang, Y. Zhang, W. Chen, CuS
4. Conclusion nanoagents for photodynamic and photothermal therapies: phenomena and
possible mechanisms, Photodiagn. Photodyn. Ther. 19 (2017) 5–14.
In summary, we successfully constructed the multifunctional [9] L. Xu, J. Liu, J. Xi, Q. Li, B. Chang, X. Duan, G. Wang, S. Wang, Z. Wang, L. Wang,
Synergized multimodal therapy for safe and effective reversal of cancer mul-
nanoparticle-integrated DMNs (CPT-CuS-ZIF-8@HA@MN) for syn- tidrug resistance based on low-level photothermal and photodynamic effects,
ergetic chemo-photothermal therapy against melanoma. Compre- Small (2018) 1800785.
hensive studies revealed that this system possessed the following [10] G. Gao, Y.W. Jiang, Y. Guo, H.R. Jia, X. Cheng, Y. Deng, X.W. Yu, Y.X. Zhu,
H.Y. Guo, W. Sun, X. Liu, J. Zhao, S. Yang, Z.W. Yu, F.M.S. Raya, G. Liang, F.G. Wu,
unique advantages: local drug delivery, improved cellular internal- Enzyme-mediated tumor starvation and phototherapy enhance mild-tempera-
ization, intelligent and spatiotemporal drug release, and synergistic ture photothermal therapy, Adv. Funct. Mater. 30 (2020) 1909391.
combination of chemotherapy and PTT. In addition, through in vivo [11] K. Poudel, A. Banstola, M. Gautam, Z. Soe, C.D. Phung, L.M. Pham, J.H. Jeong,
H.G. Choi, S.K. Ku, T.H. Tran, C.S. Yong, J.O. Kim, Macrophage-membrane-cam-
biodistribution and penetration studies, we found that the drug ouflaged disintegrable and excretable nanoconstruct for deep tumor penetra-
could be enriched at the tumor site with long duration and deliv- tion, ACS Appl. Mater. Interfaces 12 (2020) 56767–56781.
ered to deep tumor effectively after DMNs administration, which [12] K. Poudel, A. Banstola, M. Gautam, Z.C. Soe, L.M. Pham, J.H. Jeong, H.G. Choi,
S.K. Ku, C.S. Yong, T.H. Tran, J.O. Kim, Redox/photo dual-responsive, self-tar-
indicated that this DDS promisingly realized a low-dose adminis-
geted, and photosensitizer-laden bismuth sulfide nanourchins for combination
tration with high efficiency in regional application. Furthermore, therapy in cancer, Nanoscale 13 (2021) 1231–1247.
the superior tumor inhibiting performance and excellent system- [13] L. Liang, S.W. Peng, Z.W. Yuan, C. Wei, Y.Y. He, J.R. Zheng, Y.Q. Gu, H.Y. Chen,
Biocompatible tumor-targeting nanocomposites based on CuS for tumor imag-
atic safety of the DDS were confirmed in vitro and in vivo. To-
ing and photothermal therapy, RSC Adv. 8 (2018) 6013–6026.
gether, this smart design overcame the drawbacks of monotherapy [14] R. van der Meel, E. Sulheim, Y. Shi, F. Kiessling, W.J.M. Mulder, T. Lammers,
and systematic administration. Nevertheless, there are some issues Smart cancer nanomedicine, Nat. Nanotechnol. 14 (2019) 1007–1017.
need to be addressed before further utilization. For example, more [15] D.J. Irvine, E.L. Dane, Enhancing cancer immunotherapy with nanomedicine,
Nat. Rev. Immunol. 20 (2020) 321–334.
pharmacodynamics and long-term safety need to be further eval- [16] R. Wang, Z. He, P. Cai, Y. Zhao, L. Gao, W. Yang, Y. Zhao, X. Gao, F. Gao, Sur-
uated, scalable aseptic manufacturing and storage technology, and face-functionalized modified copper sulfide nanoparticles enhance checkpoint
standardized administration apparatus of the DMNs should be ex- blockade tumor immunotherapy by photothermal therapy and antigen captur-
ing, ACS Appl. Mater. Interfaces 11 (2019) 13964–13972.
ploited for further practical applications. We believe that the mul- [17] W.H. Chen, G.F. Luo, X.Z. Zhang, Recent advances in subcellular targeted cancer
tifunctional nanoparticle-integrated DMNs will be in prospect of therapy based on functional materials, Adv. Mater. 31 (2019) 1802725.
clinical application for more superficial tumor therapy with further [18] P. Majumder, U. Baxa, S.T.R. Walsh, J.P. Schneider, Design of a multicompart-
ment hydrogel that facilitates time-resolved delivery of combination therapy
delicate optimization. and synergized killing of glioblastoma, Angew. Chem. Int. Ed. Engl. 57 (2018)
15040–15044.
[19] L. Yang, H. Sun, Y. Liu, W. Hou, Y. Yang, R. Cai, C. Cui, P. Zhang, X. Pan, X. Li,
Declaration of Competing Interest
L. Li, B.S. Sumerlin, W. Tan, Self-assembled aptamer-grafted hyperbranched
polymer nanocarrier for targeted and photoresponsive drug delivery, Angew.
The authors declare that they have no known competing finan- Chem. Int. Ed. Engl. 57 (2018) 17048–17052.
cial interests or personal relationships that could have appeared to [20] J. Jiang, Y. Zhao, O.M. Yaghi, Covalent chemistry beyond molecules, J. Am.
Chem. Soc. 138 (2016) 3255–3265.
influence the work reported in this paper. [21] M. Bosch, S. Yuan, W. Rutledge, H.C. Zhou, Stepwise synthesis of metal-organic
frameworks, Acc. Chem. Res. 50 (2017) 857–865.
[22] C.Y. Sun, C. Qin, C.G. Wang, Z.M. Su, S. Wang, X.L. Wang, G.S. Yang, K.Z. Shao,
Acknowledgments Y.Q. Lan, E.B. Wang, Chiral nanoporous metal-organic frameworks with high
porosity as materials for drug delivery, Adv. Mater. 23 (2011) 5629–5632.
This work was supported by the National Natural Science [23] L. Gao, Q. Chen, T. Gong, J. Liu, C. Li, Recent advancement of imidazolate frame-
work (ZIF-8) based nanoformulations for synergistic tumor therapy, Nanoscale
Foundation (No. 81803466), the Key Areas Research and Devel-
11 (2019) 21030–21045.
opment Program of Guangdong Province (No. 2019B020204002), [24] J.C. Yang, Y. Chen, Y.H. Li, X.B. Yin, Magnetic resonance imaging-guided multi–
the Natural Science Foundation of Guangdong Province (No. drug chemotherapy and photothermal synergistic therapy with pH and NIR-s-
2021A1515012525), and Guangdong Macao joint innovation fund- timulation release, ACS Appl. Mater. Interfaces 9 (2017) 22278–22288.
[25] C.Y. Sun, C. Qin, X.L. Wang, G.S. Yang, K.Z. Shao, Y.Q. Lan, Z.M. Su, P. Huang,
ing project (No. 2020A050515009). C.G. Wang, E.B. Wang, Zeolitic Imidazolate framework-8 as efficient pH-sensi-
tive drug delivery vehicle, Dalton Trans. 41 (2012) 6906–6909.
[26] J. Feng, W. Yu, Z. Xu, J. Hu, J. Liu, F. Wang, Multifunctional siRNA-laden
Supplementary materials hybrid nanoplatform for noninvasive PA/IR dual-modal imaging-guided en-
hanced photogenetherapy, ACS Appl. Mater. Interfaces 12 (2020) 22613–
Supplementary material associated with this article can be 22623.
[27] L. He, G. Huang, H. Liu, C. Sang, X. Liu, T. Chen, Highly bioactive zeolitic imi-
found, in the online version, at doi:10.1016/j.actbio.2021.09.009. dazolate framework-8-capped nanotherapeutics for efficient reversal of reper-
fusion-induced injury in ischemic stroke, Sci. Adv. 6 (2020) eaay9751.
References [28] K. Dong, Y. Zhang, L. Zhang, Z. Wang, J. Ren, X. Qu, Facile preparation of met-
al-organic frameworks-based hydrophobic anticancer drug delivery nanoplat-
[1] M.R. Donaldson, B.M. Coldiron, No end in sight: the skin cancer epidemic con- form for targeted and enhanced cancer treatment, Talanta 194 (2019) 703–708.
tinues, Semin. Cutan. Med. Surg. 30 (2011) 3–5. [29] B. Wu, J. Fu, Y. Zhou, S. Luo, Y. Zhao, G. Quan, X. Pan, C. Wu, Tailored coreshell
[2] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, CA Cancer J. Clin. 70 (2020) dual metal-organic frameworks as a versatile nanomotor for effective synergis-
7–30. tic antitumor therapy, Acta Pharm. Sin. B 10 (2020) 2198–2211.
[3] P. Corrie, M. Hategan, K. Fife, C. Parkinson, Management of melanoma, Br. Med. [30] C. Underhill, CD44: the hyaluronan receptor, J. Cell Sci. 103 (1992) 293–298.
Bull. 111 (2014) 149–162. [31] Z. Luo, Y. Dai, H. Gao, Development and application of hyaluronic acid in tumor
[4] Y. Yan, S. Cao, X. Liu, S.M. Harrington, W.E. Bindeman, A.A. Adjei, J.S. Jang, targeting drug delivery, Acta Pharm. Sin. B 9 (2019) 1099–1112.
J. Jen, Y. Li, P. Chanana, A.S. Mansfield, S.S. Park, S.N. Markovic, R.S. Dronca, [32] H.Y. Seok, N. Sanoj Rejinold, K.M. Lekshmi, K. Cherukula, I.K. Park, Y.C. Kim,
H. Dong, CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that with- CD44 targeting biocompatible and biodegradable hyaluronic acid cross-linked
stand chemotherapy during cancer chemoimmunotherapy, JCI Insight 3 (2018) zein nanogels for curcumin delivery to cancer cells: in vitro and in vivo evalu-
e97828. ation, J. Controll. Release 280 (2018) 20–30.

177
Y. Zhao, Y. Zhou, D. Yang et al. Acta Biomaterialia 135 (2021) 164–178

[33] Z.X. Chen, M.D. Liu, M.K. Zhang, S.B. Wang, L. Xu, C.X. Li, F. Gao, B.R. Xie, [45] X. Zhang, L. Liu, L. Huang, W. Zhang, R. Wang, T. Yue, J. Sun, G. Li, J. Wang,
Z.L. Zhong, X.Z. Zhang, Interfering with lactate-fueled respiration for enhanced The highly efficient elimination of intracellular bacteria via a metal organic
photodynamic tumor therapy by a porphyrinic MOF nanoplatform, Adv. Funct. framework (MOF)-based three-in-one delivery system, Nanoscale 11 (2019)
Mater. 28 (2018) 1803498. 9468–9477.
[34] O. Gotov, G. Battogtokh, Y.T. Ko, Docetaxel-loaded hyaluronic acid-cathepsin B– [46] Q. Sun, H. Bi, Z. Wang, C. Li, X. Wang, J. Xu, H. Zhu, R. Zhao, F. He, S. Gai,
cleavable-peptide-gold nanoparticles for the treatment of cancer, Mol. Pharm. P. Yang, Hyaluronic acid-targeted and pH-responsive drug delivery system
15 (2018) 4668–4676. based on metal-organic frameworks for efficient antitumor therapy, Biomate-
[35] K. Poudel, A. Banstola, T.H. Tran, R.K. Thapa, M. Gautam, W. Ou, L.M. Pham, rials 223 (2019) 119473.
S. Maharjan, J.H. Jeong, S.K. Ku, H.G. Choi, C.S. Yong, J.O. Kim, Hyaluronic acid [47] A. Hou, G. Quan, B. Yang, C. Lu, M. Chen, D. Yang, L. Wang, H. Liu, X. Pan,
wreathed, trio-stimuli receptive and on-demand triggerable nanoconstruct for C. Wu, Rational design of rapidly separating dissolving microneedles for pre-
anchored combinatorial cancer therapy, Carbohydr. Polym. 249 (2020) 116815. cise drug delivery by balancing the mechanical performance and disintegration
[36] C. Wang, Y. Ye, G.M. Hochu, H. Sadeghifar, Z. Gu, Enhanced cancer im- rate, Adv. Healthc. Mater. 8 (2019) 1900898.
munotherapy by microneedle patch-assisted delivery of anti-PD1 antibody, [48] Y. Wang, X. Liu, G. Deng, J. Sun, H. Yuan, Q. Li, Q. Wang, J. Lu, Se@SiO2 -FA-CuS
Nano Lett. 16 (2016) 2334–2340. nanocomposites for targeted delivery of DOX and nano selenium in synergis-
[37] X. Lan, J. She, D.A. Lin, Y. Xu, X. Li, W.F. Yang, V.W.Y. Lui, L. Jin, X. Xie, tic combination of chemo-photothermal therapy, Nanoscale 10 (2018) 2866–
Y.X. Su, Microneedle-mediated delivery of lipid-coated cisplatin nanoparticles 2875.
for efficient and safe cancer therapy, ACS Appl. Mater. Interfaces 10 (2018) [49] M. Zhang, X. Liu, Q. Luo, Q. Wang, L. Zhao, G. Deng, R. Ge, L. Zhang, J. Hu,
33060–33069. J. Lu, Tumor environment responsive degradable CuS@mSiO2 @MnO2 /DOX for
[38] A.F. Moreira, C.F. Rodrigues, T.A. Jacinto, S.P. Miguel, E.C. Costa, I.J. Correia, Mi- MRI guided synergistic chemo-photothermal therapy and chemodynamic ther-
croneedle-based delivery devices for cancer therapy: a review, Pharmacol. Res. apy, Chem. Eng. J. 389 (2020) 124450.
148 (2019) 104438. [50] H. Zhang, Q. Li, R. Liu, X. Zhang, Z. Li, Y. Luan, A versatile prodrug strategy
[39] G. Mojeiko, M. de Brito, G.C. Salata, L.B. Lopes, Combination of microneedles to in situ encapsulate drugs in MOF nanocarriers: A case of cytarabine-IR820
and microemulsions to increase celecoxib topical delivery for potential appli- prodrug encapsulated ZIF-8 toward chemo-photothermal therapy, Adv. Funct.
cation in chemoprevention of breast cancer, Int. J. Pharm. 560 (2019) 365–376. Mater. 28 (2018) 1802830.
[40] A.S. Rzhevskiy, T.R.R. Singh, R.F. Donnelly, Y.G. Anissimov, Microneedles as the [51] M. Chen, D. Yang, Y. Sun, T. Liu, W. Wang, J. Fu, Q. Wang, X. Bai, G. Quan,
technique of drug delivery enhancement in diverse organs and tissues, J. Con- X. Pan, C. Wu, In situ self-assembly nanomicelle microneedles for enhanced
troll. Release 270 (2018) 184–202. photoimmunotherapy via autophagy regulation strategy, ACS Nano 15 (2021)
[41] M. Chen, G. Quan, Y. Sun, D. Yang, X. Pan, C. Wu, Nanoparticles-encapsulated 3387–3401.
polymeric microneedles for transdermal drug delivery, J. Controll. Release 325 [52] K.F. Chu, D.E. Dupuy, Thermal ablation of tumours: biological mechanisms and
(2020) 163–175. advances in therapy, Nat. Rev. Cancer 14 (2014) 199–208.
[42] W. Qin, G. Quan, Y. Sun, M. Chen, P. Yang, D. Feng, T. Wen, X. Hu, X. Pan, [53] M. Nikfarjam, V. Muralidharan, C. Christophi, Mechanisms of focal heat de-
C. Wu, Dissolving microneedles with spatiotemporally controlled pulsatile re- struction of liver tumors, J. Surg. Res. 127 (2005) 208–223.
lease nanosystem for synergistic chemo-photothermal therapy of melanoma, [54] L.F. Fajardo, B. Egbert, J. Marmor, G.M. Hahn, Effects of hyperthermia in a ma-
Theranostics 10 (2020) 8179–8196. lignant tumor, Cancer 45 (1980) 613–623.
[43] Z. Wang, X. Tang, X. Wang, D. Yang, C. Yang, Y. Lou, J. Chen, N. He, Near-in- [55] L. Wang, T. Zhang, M. Huo, J. Guo, Y. Chen, H. Xu, Construction of nucleus-tar-
frared light-induced dissociation of zeolitic imidazole framework-8 (ZIF-8) geting iridium nanocrystals for photonic hyperthermia-synergized cancer ra-
with encapsulated CuS nanoparticles and their application as a therapeutic diotherapy, Small 15 (2019) e1903254.
nanoplatform, Chem. Commun. 52 (2016) 12210–12213. [56] J. Wan, L. Huang, J. Cheng, H. Qi, J. Jin, H. Wang, Balancing the stability and
[44] J. Zhuang, C.H. Kuo, L.Y. Chou, D.Y. Liu, E. Weerapana, C.K. Tsung, Optimized drug activation in adaptive nanoparticles potentiates chemotherapy in mul-
metal-organic-framework nanospheres for drug delivery: evaluation of small– tidrug-resistant cancer, Theranostics 11 (2021) 4137–4154.
molecule encapsulation, ACS Nano 8 (2014) 2812–2819.

178

You might also like