You are on page 1of 13

Journal of Science: Advanced Materials and Devices 7 (2022) 100507

Contents lists available at ScienceDirect

Journal of Science: Advanced Materials and Devices


journal homepage: www.elsevier.com/locate/jsamd

Original Article

An investigation of IRMOF-16 as a pH-responsive drug delivery carrier


of curcumin
Mengru Cai a, 1, Boran Ni b, 1, Xueling Hu a, Kaixin Wang a, Dongge Yin a, Gongsen Chen c,
Tingting Fu a, Rongyue Zhu a, Xiaoxv Dong a, *, Changhai Qu a, **, Xingbin Yin a, ***
a
School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
b
Department of Gynaecology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
c
Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China

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

Article history: Metaleorganic frameworks (MOFs) with tunable structures, high porosity, and abundant active sites are
Received 12 May 2022 considered to be promising drug delivery systems (DDSs). In this study, novel MOFs-based nanoparticles
Received in revised form (CUR@IRMOF-16) were successfully prepared using Zn-based IRMOF-16 as a carrier and curcumin (CUR)
2 September 2022
as the model drug. This nanocargo delivery platform has dual capabilities, enabling simultaneous drug
Accepted 13 September 2022
Available online 28 September 2022
delivery and fluorescence imaging. In vitro release experiments showed that nanoparticles were released
more quickly under mildly acidic conditions, which proved their potential for tumor microenvironment-
responsive drug release. Through in vitro cell experiments, it was found that the nano-drug platform had
Keywords:
Metal-organic frameworks
high antitumor cytotoxicity, which may be related to the mechanism of increasing intracellular reactive
Drug delivery oxygen species (ROS), reducing intracellular mitochondrial membrane potential (MMP), and inducing
Curcumin apoptosis. In addition, confocal laser microscopy showed that CUR@IRMOF-16 could localize to the
pH-responsive release nucleus to exert an antitumor effect. Meanwhile, CUR@IRMOF-16 exhibited superior antitumor activity
compared with pure CUR in vitro experiments. The biocompatibility test of IRMOF-16 showed reasonable
biosafety, and no evidence of obvious toxicity was observed in vitro. CUR@IRMOF-16 has unique ad-
vantages with regard to pH response, biocompatibility, and antitumor efficacy, indicating that the nano-
drug platform is a promising drug delivery carrier with an antitumor effect.
© 2022 Vietnam National University, Hanoi. Published by Elsevier B.V. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction composed of organic acid ligands and metal cluster Zn4O [7]. MOF-5
was first reported by Yaghi in 1999, with the characteristics of high
Metaleorganic frameworks (MOFs) are nanoporous polymeric specific surface area, good thermal stability, large pore volume,
materials that are self-assembled from metal ion clusters and regular pore structure, and good hydrogen storage performance
organic ligands [1]. Recently, MOFs have been widely used for drug [8,9]. To further enhance applications of IRMOFs, Yaghi et al. ob-
delivery due to their potential application advantages, such as tained IRMOFs with different pore volumes by changing the chain
structural chemical diversity, high drug loading capacity, and good lengths of the organic ligands, such as IRMOF-8, IRMOF-10, IRMOF-
biodegradability [2e6]. One of the main challenges of biomedicine 12, IRMOF-14, and IRMOF-16 [10,11]. Interestingly, IRMOF-12
is to select appropriate carriers to effectively deliver drugs into exhibited more efficient hydrogen storage than MOF-5.
the body. Isoreticular metaleorganic frameworks (IRMOFs) are The applications of MOFs as promising drug delivery carriers
provide us with new research ideas. Currently, the application of
MOFs in the drug delivery proves its feasibility in drug packaging
[12e15]. However, there is still limited research on the effect of
* Corresponding author. expanded pore size on drug delivery. Inspired by the above, IRMOF-
** Corresponding author.
16, with the largest pore volume, has been used in drug delivery
*** Corresponding author.
E-mail addresses: 201801020@bucm.edu.cn (X. Dong), quchanghai@bucm.edu.
research. Compared with MOF-5, IRMOF-16 has longer side chains,
cn (C. Qu), yxbtcm@bucm.edu.cn (X. Yin). higher specific surface area, and larger pore volume. This study
Peer review under responsibility of Vietnam National University, Hanoi. selected IRMOF-16 as a drug delivery vehicle for the first time.
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.jsamd.2022.100507
2468-2179/© 2022 Vietnam National University, Hanoi. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/
licenses/by-nc-nd/4.0/).
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

IRMOF-16 is composed of Zn4O metal ion clusters and terphenyl- 2.2. Instrument
4,400 -dicarboxylic acid. The current research on IRMOF-16 mainly
focuses on its gas storage and release functions. Studies have SEM images were taken using a high-performance field-
explored the possibility of HO-modified IRMOF-16 under simulated emission scanning electron microscope (Zeiss, Oberkochen, Ger-
conditions and theoretically speculated on its use as a drug delivery many). 1 mg of the sample was dispersed in 5 mL of methanol and
vehicle [16e21]. sonicated for 30 min. Then the suspended droplets were placed on
Clinical applications of therapeutic drugs have been limited due to silicon wafers, and then gold was sprayed on the sample surface
inherent limitations such as poor physiological stability, nonspecific for 10 min. The Fourier transform infrared (FTIR) spectroscopy was
targeting, and low cell membrane permeability [22,23]. In many performed in the range of 4000-400 cm1 using an FTIR spec-
cases, nonspecific drug release of chemotherapeutics could cause trometer (Thermo Scientific, Waltham, USA). The samples were
damage to normal tissues and poor pharmacokinetics. Nanomaterials weighed to about 10 mg and then dispersed with KBr under dry
can control drug release rate and promote cell internalization to conditions. The X-ray diffraction (XRD) was measured utilizing an
deliver drugs. Therefore, Nanomaterials could overcome the limita- X-ray powder diffraction instrument (Nippon Science Group
tions of chemotherapeutic drugs. CUR is a natural organic chemical Corporation, Tokyo, Japan) in the range of 3e50 at a scanning
with a 1,7-diarylheptane structure, which comes from the rhizome of range of 2q using Cu-Ka (l ¼ 1.541 nm) radiation at 40 Kv and
Curcuma Longa L [24]. Modern research has shown that CUR has 40 mA. The thermogravimetric analysis (TGA) curves were recor-
special properties such as antibacterial, anti-inflammatory, anti- ded using DTG-60/60 simultaneous thermogravimetry/differen-
tumor, and antioxidant effects [25,26]. However, poor water solubility tial thermal analyzers (SHIMADZU, Kyoto, Japan) from 30  C to
and low bioavailability of CUR significantly limit its clinical applica- 600  C at a rate of 10  C/min under N2 flow. The particle size was
tions [27]. Many efforts have been made to improve the inherent determined using a Malvern Nano Zetasizer S90 (Malvern, Melvin,
limitations of CUR and increase its clinical value [28e31]. However, US). 10 mg of the samples were dispersed in 10 mL anhydrous
there is still a need to develop a DDS that can release CUR continu- ethanol and treated with ultrasound for half an hour. The surface
ously and responsively at a tumor site. area and pore size of samples were measured by nitrogen
In this study, the rapid precipitation method was used to obtain adsorption/desorption at 77 K using an ASAP 2460 analyzer
nanoscale IRMOF-16, which was used as a delivery carrier for CUR (Micromeritics, Atlanta, USA). Samples were degassed at 150  C
because of its significant biodegradability and the size of its aperture for 12 h before detection.
that could accommodate the long-chain structure of CUR. The size,
surface morphology, and drug loading performance of the synthe- 2.3. Synthesis of IRMOF-16
sized CUR@IRMOF-16 were characterized by scanning electron mi-
croscopy (SEM), Fourier transform infrared (FTIR) spectroscopy, IRMOF-16 was synthesized according to the fast precipitation
powder X-ray diffraction pattern (PXRD), thermogravimetric analysis method [32]. P-triphenyl-4,400 -dicarboxylic acid (0.06364 g) and
(TGA), dynamic light scattering (DLS), BrunauereEmmetteTeller zinc nitrate hexahydrate (Zn(NO3)2$6H2O) (0.12519 g) were dis-
(BET), and high-performance liquid chromatography (HPLC). In vitro solved in 350 mL of N,N-dimethylformamide (DMF). Then, trie-
cell experiments and confocal laser scanning microscopy (CLSM) thylamine (TEA) was added quickly while stirring at 600 rpm on a
confirmed its efficacy and toxicity against HepG2 cells. Apoptosis, multi-point intelligent magnetic stirrer for 2 h (Henan Abbot Sci-
detection of ROS, and MMP experiments were conducted to confirm ence and Technology Development Co., Ltd., Zhengzhou, China).
its antitumor-related mechanisms. The reaction liquid was centrifuged in a centrifuge (Shanghai
Fishier Analytical Instrument Co., Ltd., Shanghai, China). After
2. Experimental soaking in trichloromethane for three days, the sediment was
placed in a vacuum drying oven at 120  C for vacuum drying
2.1. Material (vacuum degree 0.01 MPa) for 12 h to activation. IRMOF-16 was
stored in a desiccator.
The zinc nitrate hexahydrate was purchased from Shanghai
Aladdin Bio-Chem Technology Co., Ltd. (Shanghai, China); P-ter- 2.4. Drug loading
phenyl-4,400 -dicarboxylic acid was acquired from Shanghai Kaishu
Chemical Technology Co., Ltd. (Shanghai, China); triethylamine The drug loading experiment was conducted following the
(TEA) was purchased from Tianjin Fuchen Chemical Reagent Factory solvent adsorption method in methanol. Three factors: the drug
(Tianjin, China); chloroform was purchased from Beijing Chemical loading times (6 h, 12 h, and 24 h), the mass ratios of MOF to CUR
Plant (Beijing, China); methanol and acetonitrile were obtained (3:2, 1:1, and 2:3), and the concentrations of CUR (1 mg/mL, 2 mg/
from Thermo Fisher Technology Co., Ltd. (Waltham, USA); dimethyl mL, and 3 mg/mL) were determined using L9(34) orthogonal tests.
sulfoxide (DMSO), EDTA-pancreatin, and DMEM were purchased The supernatant obtained in the above experiment was diluted, and
from Sigma Company (St. Louis, USA); the penicillin-streptomycin the drug concentration was evaluated using a Thermo Scientific
mixture and phosphate buffer saline (PBS) were obtained from UltiMate 3000 HPLC system (Waltham, MA, USA). The mobile phase
Beijing Solab Technology Co., Ltd. (Beijing, China); fetal bovine was a mixture of acetonitrile and water containing 4% acetic acid
serum was obtained from Zhejiang Tianhang Biotechnology Co., Ltd. (48:52), the column temperature was 30  C, the flow rate was
(Huzhou, China); curcumin (CUR) was purchased from Shanghai 1.0 mL/min, and the detection wavelength was 430 nm. The drug
Yuanye Co., Ltd. (Shanghai, China); Annexin V-FITC apoptosis loading capacity and drug loading effiency were calculated ac-
detection kit, mitochondrial membrane potential assay kit with JC-1, cording to the formula below.
and reactive oxygen species assay kit were all from Biyuntian
Biotechnology (Shanghai, China); 3-(4,5-dimethylthiazol-2-yl)-2,5- weight of drug 2 CUR @ IRMOF  16
drug loading capacity ðDLCÞ¼
dipheny-ltetrazolium bromide (MTT) was obtained from Beijing weight of CUR @ IRMOF  16
Biodee Biotechnology Co., Ltd. (Beijing, China); 40 ,6-diamidino-2-  100%
phenylindole (DAPI) was purchased from Shanghai Beyotime
(1)
Biotechnology Co., Ltd. (Shanghai, China). All chemicals were of
analytical grade or higher.
2
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

and CUR@IRMOF-16 (CUR: 9 mg/mL) for 24 h. The culture medium


weight of drug 2CUR@IRMOF 16 was sucked out from each well, and 4% tissue fixative solution was
drug loading efficiencyðDLEÞ¼
weight of CUR fixed for 10 min, and then the cells were gently washed with PBS
100% twice. The DAPI solution (200 mL, 10 mg/mL) was added to each well.
(2) Cells were stained for 10 min in dark conditions and washed twice
with PBS. The nucleus morphology was observed under a Nikon
The CUR@IRMOF-16 was dried and stored in a dryer for subse- Research Inverted Microscope ECLIPSE Ts2R (Nikon, Tokyo, Japan)
quent experiments. and photographed randomly.

2.5. In vitro drug release


2.9. Cell apoptosis
The in vitro release of CUR was studied following the dialysis
method [33]. Typically, 5 mg of CUR@IRMOF-16 was put into the The HepG2 cells were cultured into 6-well plates with 4  105
dialysis bag (the cut-off size of the dialysis bags was 8000e14000), cells/well and inoculated for 12 h, and treated with IRMOF-16
drowned in PBS with 5% tween 80 medium at pH 5.5 and pH 7.4, (IRMOF-16: 35 mg/mL) and CUR@IRMOF-16 (CUR: 9, 18 mg/mL) for
shaking at 100 rpm at 37  C. At the specified time point, 1 mL of CUR 24 h. The collected cells were washed with PBS twice. Then, 295 mL
solution was taken out, and an equal amount of fresh buffer was Annexin V-FITC binding solution, 5 mL annexin V-FITC staining so-
replenished. The concentration of CUR was determined by HPLC. lution, and 10 mL PI staining solution were added successively. After
The concentrations were calculated by interpolation from the incubating for 20 min in the dark, flow detection was performed.
calibration curves using linear regression models. The CUR cali-
bration curves were linear over the concentration range from
2.10. Mitochondrial membrane potential (MMP)
0.00004 mg/mL to 0.7 mg/mL (y ¼ 1481.3x - 3.4549, R2 ¼ 0.9999,
Table S6 and Fig. S3). The cumulative release rate was calculated
The HepG2 cells were cultured in 12-well plates with a density
using the formulae below:
of 2  105 cells/well and inoculated for 12 h, and then treated with
IRMOF-16 (IRMOF-16: 35 mg/mL), CUR, and CUR@IRMOF-16 (CUR:
n X
t 1
9 mg/mL) for 24 h. After removing the medium, the cells were
CC ¼ Ct þ Ct (3)
V 0 washed gently with PBS. The JC-1 working solution was added to
each well for 30 min, and then the cells were washed twice with JC-
MR 1 buffer solution in the ice bath. The cells were observed under a
Drug release ¼  100% (4) fluorescence inverted microscope.
ML
Cc represented the adjusted concentration of CUR at time t, Ct
was the measured concentration at t, v was the volume of the 2.11. Cellular uptake experiment
derived samples, and V was the volume of release solution. MR and
ML were the amounts of released drug and loaded drug [equation The HepG2 cells were inoculated in a laser confocal dish at a
(4)], respectively. The zero-order drug release equation, first-order density of 1  105 cells/well and cultured for 12 h, and then CUR
drug release equation, Higuchi equation, and RitgerePeppas and CUR@IRMOF-16 (CUR: 9 mg/mL) were added for 24 h. The
equation were used to fit the drug release properties of culture medium was sucked out, and the cells were washed twice
CUR@IRMOF-16 in vitro. with PBS, followed by lysosome staining with LysoTracker Red for
20 min and nuclear staining with DAPI solution. After washing with
2.6. Cell culture PBS three times, 1 mL PBS was added for observation under a laser
confocal microscope.
Guangzhou Jeniobio Biotechnology provided the HepG2 cells.
The cells were cultured in a high-glucose DMEM containing 10%
fetal bovine serum (FBS) and 1% penicillin-streptomycin solution at 2.12. Reactive oxygen species (ROS)
37  C in a 5% CO2 incubator.
The intracellular ROS level was measured using a reactive oxy-
gen species assay kit (Beyotime Biotechnology, Shanghai, China).
2.7. Cell viability
The HepG2 cells were seeded with 3  105 cell/well in 6-well plates
and grew for 12 h, and then treated with CUR (CUR: 9 mg/mL) and
The in vitro cytotoxicity of IRMOF-16 and CUR@IRMOF-16 to
CUR@IRMOF-16 (CUR: 9 mg/mL) for 24 h. The medium was dis-
HepG2 cells was evaluated using MTT assays. The HepG2 cells were
carded, and the DCFH-DA working solution diluted with serum-free
seeded into 96-well plates with 8000 cells/well, and treated with
medium (1.2 : 1000) was replaced for 20 min. The DCFH-DA was
different concentrations of IRMOF-16 (IRMOF-16: 5, 10, 15, 20, 25, 30,
oxidized to DCF by ROS. After washing 3 times, observations were
35 mg/mL) and CUR@IRMOF-16 (CUR: 3, 6, 9, 12, 15, 18 mg/mL) for 24 h
carried out under an inverted fluorescence microscope at 488 nm
and 48 h. The MTT solution was added to each well (20 mL, 5 mg/mL)
and 525 nm.
and then incubated at 37  C for 4 h. The medium was removed, and
150 mL DMSO was added to each well. After shaking the 96-well plate
for 10 min, the absorbance was measured at 490 nm using a 2.13. Statistical analysis
microplate reader (BioTek Instruments, Inc., Vermont, USA).
All experiments were performed with at least three indepen-
2.8. Nuclear morphology dent replications, and data were expressed as means ± SDs. Com-
parisons were analyzed using one-way variance analysis (ANOVA).
The HepG2 cells were cultured with 1  105 cells/well in 24-well *p < 0.05 and ***p < 0.001 were considered to be statistically
plates and then treated with IRMOF-16 (IRMOF-16: 5, 15, 35 mg/mL) significant.
3
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

3. Results addition, the peak at 2200 cm1 may be contributed by NeC]O in


the residual DMF. The XRD pattern of IRMOF-16 was compared
3.1. Synthesis and characterization with the simulated PXRD pattern of an original IRMOF-16 crystal
[37]. As shown in Fig. 2a, the peak positions of the synthesized
The synthesized IRMOF-16 was obtained according to the rapid IRMOF-16 are in general agreement with the simulated pattern of
precipitation method and characterized by SEM, TEM, XRD, FTIR, IRMOF-16 [37]. The FTIR and XRD results indicated that IRMOF-16
TGA, and DLS [34]. The SEM and TEM image shows the synthesized was successfully synthesized. The TGA curve of IRMOF-16 is
IRMOF-16 were smooth and spherical (Fig. 1a and c). The FTIR shown in Fig. 2b. The small weight loss at about 100  C might be
image (Fig. 2d) indicates that IRMOF-16 was successfully synthe- caused by the evaporation of absorbed water in the skeleton [38].
sized. The absorption peaks are at 826 cm1 and 749 cm1, which The weight loss of the skeleton was obvious when the temperature
are caused by different types of CeH bonds in the phenyl group that was around 300  C, indicating that IRMOF-16 could maintain the
demonstrate the presence of terphenyl-4,400 -dicarboxylic acid and framework structure and had good thermal stability below 300  C.
the formation of the structural framework of IRMOF-16 [35]. The It can be observed from Fig. S1a that the particle size of IRMOF-16 is
peak at 522 cm1 is the absorption peak of the ZneO bond in the about 284.6 nm, which is consistent with the SEM result. The sharp
Zn4O tetrahedral metal cluster [36]. The peaks at 1700e1380 cm1 peak reveals that the particle size of IRMOF-16 is relatively
are mainly characteristic CeO vibration peaks. The CeO vibration concentrated, which indicates that the synthesized IRMOF-16 has a
peaks of the carboxylic acid functional groups are mainly located in small particle size difference and is suitable for drug loading
this region. The peak at 1590 cm1 is the asymmetric stretching experiments. Fig. 2c shows the result of the N2
vibration of the COO bond in terphenyl-4,400 -dicarboxylic acid, adsorptionedesorption experiment of IRMOF-16. The Bru-
while the peak at 1394 cm1 is the symmetric stretching vibration. nauereEmmetteTeller (BET) surface area of IRMOF-16 is 94.15 m2/
The CeO bond of the carboxyl group on terphenyl-4,400 -dicarbox- g, and the total volume in pores is 0.162 cm3/g with a pore diameter
ylic acid is not broken, and the carboxyl group is bridged with the of 6.495 nm. The results show that the synthesized IRMOF-16 has a
metal cluster. The wide peak at 3500 cm1 indicates the presence of nanoporous structure, which also proves that IRMOF-16 is suitable
a small amount of weakly bound water in the material [13]. In for subsequent experiments.

Fig. 1. (a) SEM image of IRMOF-16; (b) SEM image of CUR@IRMOF-16; (c) TEM image of IRMOF-16; (d) TEM image of CUR@IRMOF-16.

4
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

Fig. 2. (a) XRD patterns of IRMOF-16 and IRMOF-16 (simulation); (b) thermogravimetric (TG) analysis of IRMOF-16, CUR, and CUR@IRMOF-16; (c) BET isotherm of IRMOF-16 and
CUR@IRMOF-16 at 77 K by N2; (d) FTIR spectra of CUR, IRMOF-16, and CUR@IRMOF-16.

3.2. Drug loading TEM, DLS, FTIR, and DTG. The nanoparticles remained spherical with
little change in particle size (Fig. 1b and d). And the DLS proved the
According to the results of the orthogonal experiment, the particle size of CUR@IRMOF-16 was 288.3 nm (Fig. S1b). Fig. 2d
optimal drug loading conditions were as follows: the drug loading shows the FT-IR spectra of IRMOF-16, CUR, and CUR@IRMOF-16. In
time was 6 h, the weight ratio of IRMOF-16 to CUR was 2:3, and the the FT-IR spectrum of IRMOF-16, the characteristic peaks at
concentration of CUR was 3 mg/mL. Under these conditions, the DLC 1590 cm1 and 1394 cm1 indicate that the COOH group of tri-
reached 65.67%, and the DLE reached 43.78% (Tables S2eS4). And the phenyl-4,400 -dicarboxylic acid is deprotonated upon reaction with
CUR@IRMOF-16 obtained under this condition has good reproduc- metal ions [41]. In the spectrum of CUR, the band at 3510 cm1
ibility (Table S5). The high drug loading capacity may be attributed to represents the OeH vibration of the phenolic group. Other charac-
the large pore size of IRMOF-16 and the high affinity of CUR and teristic peaks of the CUR are found at 1630 cm1 (C]C vibration),
IRMOF-16 [39,40]. Then, CUR@IRMOF-16 was characterized by SEM, 1602 cm1 (benzene ring stretching vibration), 1505 cm1 (C]O

Fig. 3. (a) Zeta potential of CUR and CUR@IRMOF-16; (b) drug release curve of CUR@IRMOF-16 in PBS containing 5% tween 80 at pH 7.4 and 5.5 (n ¼ 3, mean þ SD).

5
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

vibration), 1429 cm1 (olefinic CeH bending vibrations), 1280 cm1 respectively, at 100  C (Fig. S2). This may be related to the moisture
(aromatic CeO stretching vibrations), and 1029 cm1 (CeOeC contained in MOFs and drugs. And there was a slight weight loss at
stretching vibrations) [13]. The spectral comparison of CUR@IRMOF- 200  C, probably due to the volatilization of residual DMF. When it
16 and IRMOF-16 exhibits that the CUR@IRMOF-16 spectrum has exceeds 300  C, CUR begins to degrade with a large amount of
three signals at 1505 cm1, 1630 cm1, and 3530 cm1, corre- weight loss. The weight loss procedures of IRMOF-16 and
sponding to the C]O, C]C, and OeH functional groups of CUR, CUR@IRMOF-16 are relatively similar, indicating a loss of CUR crys-
respectively. The FT-IR spectra results confirm the presence of CUR in tallinity in IRMOF-16 [42]. However, the decomposition rate of
the CUR@IRMOF-16 sample. Moreover, due to several methanol CUR@IRMOF-16 was slower than that of CUR, which may be related
washings during the drug loading process, the residual DMF is also to the protective effect of IRMOF-16 on CUR. Liang et al. also
removed, so there is no 2200 cm1 peak in CUR@IRMOF-16. There- demonstrated the protective effect of ZIF-90 on the enzyme at high
fore, the drug loading process is also regarded as killing two birds temperature [43]. Liang et al. attributed this protective effect to MOF/
with one stone, which can not only successfully load CUR into biointerface, which would greatly improve the activity and stability
IRMOF-16, but also further remove the residual organic reagents in of the drug [44]. After adsorb-ing CUR, the BET surface area of
the material. As shown in Fig. 1b, IRMOF-16, CUR, and CUR@IRMOF- CUR@IRMOF-16 was 34.0035 m2/g, and the total volume in pores
16 have good thermal stability before 300  C. The weight losses of was 0.21201 cm3/g (Fig. 2a). Also, the z-potential values changed
IRMOF-16, CUR, and CUR@IRMOF-16 were 8%, 9%, and 6%, from 5.99 to 6.38 mV after CUR loading (Fig. 3a). There is little

Table 1
The fitting curve of CUR@IRMOF-16 by different mathematical models under pH values of 7.4 and 5.5.

Model Equation (0e120 h) R2 (0e120 h) Equation (120e240 h) R2 (120e240 h)

pH ¼ 5.5 Zero-order kinetic equation Mt ¼ 0.03258 þ 0.12976t 0.99537 Mt ¼ 3.96229 þ 0.0925t 0.99078
First-order kinetic equation Mt ¼ 70.91282(1-e0.00201t) 0.99709 Mt ¼ 58.48983(1-e0.00244t) 0.99419
Higuchi equation Mt ¼ 1.65995t1/2 - 4.05712 0.97872 Mt ¼ 2.45341t1/2 - 12.09704 0.99417
RitgerePeppas equation ln(Mt) ¼ 0.95671lnt-1.38276 0.99069 ln(Mt) ¼ 0.79979lnt-1.12342 0.9928
pH ¼ 7.4 Zero-order kinetic equation Mt ¼ 0.00403 þ 0.17035t 0.9984 Mt ¼ 2.33912 þ 0.14988t 0.99063
First-order kinetic equation Mt ¼ 206.77841(1-e0.0008577t) 0.99871 Mt ¼ 189.17327(1-e0.0009394t) 0.99046
Higuchi equation Mt ¼ 2.17086t1/2 - 5.31217 0.97352 Mt ¼ 3.96667t1/2 - 23.56886 0.9889
RitgerePeppas equation ln(Mt) ¼ 0.98201lnt - 1.68884 0.99852 ln(Mt) ¼ 0.91717lnt - 1.27283 0.98927

Fig. 4. (a) Cytotoxicity study of IRMOF-16 on the HepG2 cells determined by MTT assay; (b) Cytotoxicity study of IRMOF-16 on the L02 cells determined by MTT assay; (c) Apoptosis
assays for HepG2 cells after treatment with IRMOF-16 for 24 h; (d) Fluorescent microscopic images of the DAPI-stained HepG2 cells following 24 h treatment with different
concentrations of IRMOF-16.

6
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

difference in z-potential values before and after drug loading, which the in vitro release kinetics of the drug. At a pH of 5.5, CUR@IRMOF-
proves that the drug is successfully loaded into the pores of MOFs. 16 conformed to the first-order kinetic equation and exhibited a
Negative surface charges can prolong the circulation time of nano- sustained-release effect. At a pH of 7.4, CUR@IRMOF-16 conformed to
particles in vivo and avoid clearance by the reticuloendothelial sys- the first-order kinetic equations in the first stage (0e120 h) and the
tem (RES) [45]. zero-order kinetic equations in the second stage (120e240 h). The
Higuchi model is a model used to study the release mechanism. The
3.3. In vitro drug release correlation coefficients of the Higuchi equation were less than 0.99,
indicating that there was a deviation in fitting the release behavior of
To investigate the in vitro release performance of CUR@IRMOF- CUR@IRMOF-16 with this model [46]. The RitgerePappas model is a
16, two different pH values (pH 5.5 and pH 7.4) were selected. As model used to judge the matrix dissolution and drug diffusion
shown in Fig. 3b, CUR continued to be released within 240 h and behavior during drug release [47]. The results show that the co-
showed a pH-responsive drug release. CUR@IRMOF-16 NPs slowly efficients of lnt values of this model are greater than 0.89, indicating
released CUR in PBS with a pH of 7.4, and the CUR released by the that the in vitro drug release behavior of CUR@IRMOF-16 is mainly
system at the end of 240 h was less than 25%, which was beneficial caused by the dissolution of CUR@IRMOF-16. The ability of IRMOF-16
to avoid the dissipation of CUR before reaching the target. The to load CUR is as high as 65.67%, and it can release CUR continuously
sustained release of CUR@IRMOF-16 was related to the encapsu- and is Ph-responsive; therefore, CUR@IRMOF-16 shows significant
lation of IRMOF-16 by shells that played a protective role around advantages in tumor therapy [48].
the CUR. In addition, the sustained release of CUR was related to its
affinity with Zn ions. 3.4. The biosafety evaluation of IRMOF-16
The cumulative release rates of CUR@IRMOF-16 at different pH
values were fitted to the release equation to determine the release The good biosafety of NPs was the basis for its application in the
characteristics. It can be seen that CUR@IRMOF-16 has an obvious treatment process. As shown in Fig. 4a, the survival rates of the
two-stage release behavior. The first stage is the initial release from HepG2 cells incubated with IRMOF-16 for 24 and 48 h are both
0 to about 120 h. Then, the shape of the release curve changes and greater than 85%, indicating that IRMOF-16 is nontoxic to HepG2
remains different from 120 to 240 h. Separate kinetic analyses were cells at a concentration range of 35 mg/mL. Similarly, IRMOF-16 was
performed using these two distinct time ranges, and the results are safe and nontoxic to L02 cells at a concentration range of 35 ug/mL
shown in Table 1. The zero- and first-order kinetic equations describe (Fig. 4b). DAPI staining was used to observe the effect of IRMOF-16

Fig. 5. (a) Cytotoxicity study of CUR on HepG2 cells determined by MTT assay; (b) cytotoxicity study of CUR on L02 cells determined by MTT assay; (c) cytotoxicity study of
CUR@IRMOF-16 on HepG2 cells determined by MTT assay; (d) cytotoxicity study of CUR@IRMOF-16 on L02 cells determined by MTT assay; *p < 0.05 and ***p < 0.01.

7
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

on the nucleus morphology. As shown in Fig. 4d, the nucleus that CUR@IRMOF-16 had a higher survival rate compared to pure
morphology of the HepG2 cells incubated with different concen- CUR. This is related to the slow release of CUR in IRMOF-16.
trations of IRMOF-16 did not change significantly. To further verify
the biological safety of IRMOF-16, an apoptosis experiment was 3.6. Cellular uptake experiment
carried out. A certain dose of IRMOF-16 did not induce apoptosis of
HepG2 cells, which proved the safety of IRMOF-16 once again To study and determine the localization of CUR@IRMOF-16 in
(Fig. 4c). HepG2 cells, the intracellular distribution of the nano platform was
analyzed by confocal laser microscopy. The intracellular fluores-
3.5. The cytotoxicity of CUR@IRMOF-16 cence imaging results (Fig. 6a) presented that the intracellular
green fluorescence of CUR@IRMOF-16 is more obvious than that of
The antitumor efficacy of CUR@IRMOF-16 in vitro was investi- pure CUR, indicating that the cellular uptake of CUR@IRMOF-16 by
gated through the MTT experiment. The viability of HepG2 cells HepG2 cells is stronger. The translocation of CUR@IRMOF-16 in
incubated with CUR (Fig. 5a) or CUR@IRMOF-16 (Fig. 5c) was HepG2 cells was significantly higher than that of CUR, possibly due
significantly reduced. The toxicity of CUR and CUR@IRMOF-16 was to the passive targeted delivery of nano IRMOF-16 [49]. In addition,
significantly enhanced with an increase in concentration and pro- the green fluorescence is more distributed in the nucleus and
longed incubation time. Interestingly, the toxicity of CUR@IRMOF- lysosome, indicating that nanoparticles could be localized to the
16 was greater than that of CUR within 24 h, which may have nucleus and lysosome to play an antitumor role.
been related to the enhanced cellular uptake of CUR@IRMOF-16 by
HepG2. However, the toxicity of CUR was greater than that of 3.7. Reactive oxygen species (ROS)
CUR@IRMOF-16 within 48 h, which may have been due to the slow
release of CUR in CUR@IRMOF-16 after cellular uptake, and CUR CUR was shown to be a compound with excellent antitumor
directly exerted an antitumor effect. This result indicates that effect, which can effectively induce apoptosis of HepG2 cells by
CUR@IRMOF-16 has the potential to sustain its efficacy in vivo. In elevating intracellular reactive oxygen species [50,51]. Thus, the
addition, we also demonstrated the toxicity of CUR and level of ROS was detected, and the results showed that
CUR@IRMOF-16 to L02 cells (Fig. 5b and d). It was clearly observed CUR@IRMOF-16 significantly increases intracellular ROS in HepG2

Fig. 6. (a) Reactive oxygen species assays for HepG2 cells after treatment with CUR and CUR@IRMOF-16; (b) cellular uptake assays for HepG2 cells after treatment with CUR and
CUR@IRMOF-16.

8
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

(Fig. 6b). Similarly, CUR also induced the production of ROS in L02 3.9. In vivo biodistribution
cells, but the production of ROS in L02 cells by CUR@IRMOF-16 was
not obvious, which may be due to the increased uptake of In vivo biodistribution was assessed by in vivo fluorescence
CUR@IRMOF-16 by HepG2 cells due to the enhanced permeability imaging using cy5.5-labeled CUR and cy5.5-CUR@IRMOF-16
and retention (EPR) effect. The lower production of ROS in L02 cells (N ¼ 4). cy5.5-labeled CUR and cy5.5-CUR@IRMOF-16 showed
by CUR@IRMOF-16 was also related to the sustained release of CUR, different biodistributions (Fig. 8a). Systemic accumulation was
while the EPR effect enhanced the uptake of nanoparticles by observed in the cy5.5-labeled CUR group, especially in the liver,
HepG2 cells, so the production of ROS by CUR was greatly increased which may be related to hepatic metabolism. The cy5.5-labeled
in HepG2 cells. As shown in Fig. 6, the significant increase in green CUR@IRMOF-16 group showed a certain degree of tumor target-
fluorescence indicates a significant increase in intracellular ROS, ing within 1 h. The EPR effect enhanced the phagocytosis of the
which may be related to the activation of the apoptotic pathway. nanosystem by tumor tissue. cy5.5-labeled CUR@IRMOF-16 mainly
accumulated in the tumor site from 12 h. In vitro fluorescence
imaging could be used to analyze drug retention in major organs
3.8. Cell apoptosis and tumors (Fig. 8b). cy5.5-labeled CUR is distributed in various
organs, and cy5.5-labeled CUR@IRMOF-16 is mainly distributed in
The nucleus morphology was observed to further evaluate the tumor sites. EPR effect and proton sponge effect resulted in stronger
antitumor effect of CUR@IRMOF-16. Cells treated with CUR@IRMOF- accumulation of cy5.5-labeled CUR@IRMOF-16 in tumors.
16 had strong nuclear fluorescence, accompanied by nuclear pyk-
nosis and rupture, which proved that CUR@IRMOF-16 might induce
apoptosis (Fig. 7a). The membrane potential of cells was further 4. Discussion
tested to explore the effect of CUR@IRMOF-16 on mitochondria. As
shown in Fig. 7c, the enhanced green fluorescence and reduced red There are many studies on MOFs as antitumor drug delivery
fluorescence under the fluorescence microscope indicate that vehicles [52,53]. To improve drug delivery efficiency, more and
mitochondrial membrane potential is decreased and the mitochon- more MOFs have been synthesized and reported [54,55]. Currently,
dria are damaged, which may be one possible way by which drug delivery strategies based on different principles have been
CUR@IRMOF-16 induces apoptosis. Therefore, apoptosis experi- developed, including surface adsorption, pore encapsulation, co-
ments were conducted to demonstrate the possible antitumor valent binding, and using functional molecules as building blocks
mechanism of CUR@IRMOF-16. It was found that CUR@IRMOF-16 [56]. Tuning the organic ligands of MOFs is the most facile and
had the effect of inducing cell apoptosis and showed a rey's group
effective way to tune their properties. In 2006, the Fe
concentration-dependent effect at the same time (Fig. 5b). conducted the first study of MOFs as drug delivery vehicles [57].

Fig. 7. (a) fluorescent microscopic images of DAPI-stained HepG2 cells following 24 h treatment with CUR and CUR@IRMOF-16; (b) apoptosis assays for HepG2 cells after treatment
with CUR@IRMOF-16 for 24 h, **p < 0.05 and ***p < 0.01; (c) MMP assays for HepG2 cells after treatment with IRMOF-16, CUR, and CUR@IRMOF-16; *p < 0.05 and ***p < 0.01.

9
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

Fig. 8. The biodistribution of different NPs. (a) In vivo biodistribution images of cy5.5-labeled CUR and CUR@IRMOF-16 at 1, 4, 8, 12 and 24 h, (b) fluorescence distribution images of
tumors and different organs 24 h after injection.

They used iron-based metaleorganic frameworks MIL-100 and the adsorption performance, but the smaller the pore size, the
MIL-101 as porous matrices, and ibuprofen was used as a model longer the void space, which is not conducive to the diffusion of
drug for drug delivery. Each gram of MIL-101 could load 1.376 g of reactants. Macropores are too large for common reactants or
ibuprofen, which was significantly more than 0.347 g/g of MIL-100. solvents but facilitate the exposure of other pores. In the follow-
The difference in drug loading between the two was mainly due to up, the research scope of pore size is further expanded, and the
their different pore sizes, and both had good sustained-release ef- corresponding drug loading laws of materials with different pore
fects [58]. size types are discussed. The large specific surface area provides
According to previous studies, IRMOF-1 has been used as a more active sites for the adsorption of CUR. This work indicated
drug delivery carrier with CUR. When a MOF has been modu- that it was feasible to match the structure by changing pore size
lated by introducing a functional group into the structure, the using different organic ligands. In this study, the drug delivery
drug loading capacity has been higher than that of IRMOF-1 [14]. system was constructed using IRMOF-16 as the carrier for the
Studies on the influence of pore size on MOFs have been inspired first time. The high drug loading capacity of IRMOF-16 carrying
by this result. In this work, we synthesized IRMOF-16, which had CUR was due to its excellent BET surface area (94.15 m2/g) and
the same topology as IRMOF-1. In order to avoid other factors the large volume in pores (0.162 cm3/g). Dang et al. compared
affecting drug delivery as much as possible, IRMOF-16 was syn- the adsorption of different MOFs to CUR and found it was
thesized with full reference to the synthesis method of IRMOF-1 obvious that without the effects of substituent incorporation, the
[14]. IRMOF-16 was successfully synthesized with a drug loading MOFs with the higher surface area could load more CUR [13].
capacity of 65.67%. The synthesized IRMOF-16 is a mesoporous The high drug loading capacity of IRMOF-16 for CUR is also
material; that is, the pore size is in the range of 2e50 nm. As a related to its high affinity for CUR. There is a b-diketone functional
mesoporous material, IRMOF-16 has a relatively large specific group in the CUR structure, and its 1,3-diketone can be automati-
surface area and large pore size, which is more conducive to cally converted into a tautomeric form of ketoeenol, while
transporting reactants or solvents and is also conducive to the ketoeenol is more stable and can easily chelate metal ions such as
loading and release of drugs. Micro and macroporous materials Zn2þ and Cu2þ [39]. Xing et al. [40] prepared nanoparticles by
have different properties than mesoporous materials. The more utilizing the chelation between CUR and Zn2þ, which was more
micropores, the larger the specific surface area and the stronger easily released under acidic conditions due to the acid-labile

10
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

coordination bond of Zn(II)eO. However, this strategy is limited in Funding


the packet-carrying capacity of CUR. The use of MOFs for CUR de-
livery still shows great appeal. This research was funded by the Fundamental Research Funds
It was amazing that the CUR was stably released even after for the Central Universities (No. 2020-JYB-ZDGG-046), the Beijing
160 h, which showed the excellent slow-release feature of Natural Science Foundation (No. 7202121), and the National Nat-
CUR@IRMOF-16. CUR@IRMOF-16 could release more CUR under a ural Science Foundation of China (No. 81703715), Young Elite Sci-
pH of 5.5. The pH-responsive drug delivery system showed its entists Sponsorship Program by CACM (CACM-QNRC2-A03).
advantage for tumor treatment. To save time and cost, we only
conducted a 240-h re-lease experiment. The percentage of total
Declaration of competing interest
drug release did not reach 80% at 240 h; the drug may have been
located inside some micropores that needed more time [46]. Due to
The authors declare that they have no known competing
the complex action of CUR and zinc ion center, CUR@IRMOFs can be
financial interests or personal relationships that could have
released slowly at a constant speed, and the release time can last for
appeared to influence the work reported in this paper.
more than 10 days. Pore adsorption, surface adsorption, p-p
conjugation, and electrostatic adsorption can occur between MOFs
and CUR. Various forces have a significant influence on the loading Appendix A. Supplementary data
capacity and release rate of CUR. In addition, the structure and
properties of MOFs also have certain effects on their drug delivery Supplementary data to this article can be found online at
behavior, such as porosity, functionalization, size, crystallinity, and https://doi.org/10.1016/j.jsamd.2022.100507.
surface charge. In order to reduce administration frequency,
improve patient compliance, and reduce side effects, drugs are References
often designed as sustained-release preparations. Boi et al. [59]
designed and developed alginate microspheres for loading adria- [1] O. Yaghi, G. Li, H. Li, Selective binding and removal of guests in a microporous
mycin to control its release rate and reduce its toxicity. Meng et al. metal-organic framework, Nature, 378 (1995) 703e706, https://doi.org/
10.1038/378703a0.
[60] designed and developed a quaternized pectin montmorillonite [2] Joseph Della Rocca, Demin Liu, Wenbin Lin, Nanoscale metal-organic frame-
composite membrane to delay the release of 5-Fu, which could works for biomedical imaging and drug delivery, Acc. Chem. Res. 44 (2011)
improve the problem of frequent drug administration caused by 957e968, https://doi.org/10.1021/ar200028a.
[3] J. Gandara-Loe, B.E. Souza, A. Missyul, G. Giraldo, J.C. Tan, J. Silvestre-Albero,
short half-life.
MOF-based polymeric nanocomposite films as potential materials for drug
As novel drug delivery vehicles, the biosafety of MOFs deserves delivery devices in ocular therapeutics, ACS Appl. Mater. Interfaces 12 (2020)
attention. In recent years, numerous studies have reported on the 30189e30197, https://doi.org/10.1021/acsami.0c07517.
[4] Aba nades Lazaro Isabel, C.J.R. Wells, R.S. Forgan, Multivariate modulation of
biosafety of MOFs in vitro and in vivo [61e66], which have indi-
the Zr MOF UiO-66 for defect-controlled combination anticancer drug de-
cated that they have good biosafety at a certain dose. In the MTT, livery, Angew. Chem., Int. Ed. 59 (2020) 5211e5217, https://doi.org/10.1002/
DAPI staining, and the cell apoptosis assays, IRMOF-16 showed anie.201915848.
great biocompatibility since it did not affect the nucleus [5] Xiaoti Yang, Tang Qiao, Ying Jiang, Meining Zhang, Ming Wang, Lanqun Mao,
Nanoscale ATP-responsive zeolitic imidazole framework-90 as a general
morphology or induce apoptosis. After comprehensive testing for platform for cytosolic protein delivery and genome editing, J. Am. Chem. Soc.
IRMOF-16, the safe dose of IRMOF-16 for HepG2 cells reached 141 (2019) 3782e3786, https://doi.org/10.1021/jacs.8b11996.
35 mg/mL. As compared with other biological materials, the safe [6] C. Orellana-Tavra, M. Ko €ppen, A. Li, N. Stock, D. Fairen-Jimenez, Biocompatible,
crystalline, amorphous bismuth-based metal-organic frameworks for drug
concentration of IRMOF-16 is lower. However, in vitro pharmaco- delivery, ACS Appl. Mater. Interfaces 12 (2020) 5633e5641, https://doi.org/
dynamic tests found that CUR@IRMOF-16 could exert a good anti- 10.1021/acsami.9b21692.
tumor effect within the safe concentration range. As compared [7] Zehan Mai, Dingxin Liu, Synthesis and applications of isoreticular metal-
organic frameworks IRMOFs-n (n ¼ 1, 3, 6, 8), Cryst. Growth Des. 19 (2019)
with pure CUR, the toxicity of CUR@IRMOF-16 decreased in the
7439e7462, https://doi.org/10.1021/acs.cgd.9b00879.
same time frame, which was attributed to sustained-release [8] H. Li, M. Eddaoudi, M. OM Yaghi O'Keeffe, Design and synthesis of an
properties of IRMOF-16. exceptionally stable and highly porous metal-organic framework, Nature 402
(1999) 276e279, https://doi.org/10.1038/46248.
In this study, we synthesized IRMOF-16 and successfully ob-
[9] D. Saha, Z. Bao, F. Jia, S. Deng, Adsorption of CO(2), CH(4), N(2)O, and N(2) on
tained CUR@IRMOF-16. IRMOF-16 has great biodegradability and MOF-5, MOF-177, and zeolite 5A, Environ. Sci. Technol. 44 (2010) 1820e1826,
could load more CUR than other MOFs [13,14]. Researchers can https://doi.org/10.1021/es9032309.
design more drug delivery systems based on IRMOF-16. [10] Steven S. Kaye, Anne Dailly, Omar M. Yaghi, Jeffrey R. Long, Impact of prep-
aration and handling on the hydrogen storage properties of Zn4O(1,4-
benzenedicarboxylate)3 (MOF-5), J. Am. Chem. Soc. 129 (2007)
14176e14177, https://doi.org/10.1021/ja076877g.
5. Conclusions [11] H. Furukawa, K.E. Cordova, M. O'Keeffe, O.M. Yaghi, The chemistry and ap-
plications of metal-organic frameworks, Science 341 (2013), 1230444, https://
doi.org/10.1126/science.1230444.
We designed a new multifunctional drug delivery system, i.e., [12] R.C. Alves, Z.M. Schulte, M.T. Luiz, P. Bento da Silva, R.C.G. Frem, N.L. Rosi, M.
CUR@IRMOF-16, to enhance antitumor efficiency by enhancing Chorilli, Breast cancer targeting of a drug delivery system through post-
synthetic modification of curcumin@N3-bio-MOF-100 via click chemistry,
cellular uptake and inducing apoptosis. The in vitro drug release Inorg. Chem. 60 (2021) 11739e11744, https://doi.org/10.1021/acs.inorgchem.
results showed that CUR@IRMOF-16 had sustained-release ca- 1c00538.
pacity over 10 days and exhibited pH sensitivity. The experimental [13] Y Thi Dang, Minh-Huy Dinh Dang, Ngoc Xuan Dat Mai, Linh Ho Thuy Nguyen,
Thang Bach Phan, Hai Viet Le, Tan Le Hoang Doan, Room temperature syn-
cytotoxicity results indicated that CUR@IRMOF-16 had good thesis of biocompatible nano Zn-MOF for the rapid and selective adsorption of
antitumor activity. CUR@IRMOF-16 showed good intracellular curcumin, J. Sci.: Adv. Mater. Devices 5 (2020) 560e565, https://doi.org/
uptake and nuclear localization in the fluorescence imaging 10.1016/j.jsamd.2020.09.009.
[14] M. Cai, L. Qin, L. Pang, B. Ma, J. Bai, J. Liu, X. Dong, X. Yin, J. Ni, Amino-func-
analysis. Through the apoptosis experiment, it was found that
tionalized Zn metal-organic frameworks as antitumor drug curcumin carriers,
CUR@IRMOF-16 induced tumor cell apoptosis by promoting a New J. Chem. 44 (2020) 17693e17704, https://doi.org/10.1039/D0NJ03680C.
decrease in mitochondrial membrane potential. However, the [15] R.K. Alavijeh, K. Akhbari, Biocompatible MIL-101(Fe) as a smart carrier with
antitumor mechanism of CUR@IRMOF-16 needs to be further high Loa1ding potential and sustained release of curcumin, Inorg. Chem. 59
(2020) 3570e3578, https://doi.org/10.1021/acs.inorgchem.9b02756.
verified, and the in vivo antitumor effects of the drug delivery [16] Yamil J. Colon, Rajamani Krishna, Randall Q. Snurr, Strong influence of the H2
system should also be studied in further works in more detail. binding energy on the MaxwelleStefan diffusivity in NU-100, UiO-68, and

11
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

IRMOF-16, Microporous Mesoporous Mater 185 (2014) 190e196, https:// theoretical calculation, J. Mol. Struct. 984 (2010) 316e325, https://doi.org/
doi.org/10.1016/j.micromeso.2013.10.031. 10.1016/j.molstruc.2010.09.049.
[17] Ege Dundar, Justyna Rogacka, Lucyna Firlej, Carlos Wexler, Philip Llewellyn, [40] Z.H. Xing, J.H. Wei, T.Y. Cheang, Z.R. Wang, X. Zhou, S.S. Wang, W. Chen,
Boulet Pascal, Bogdan Kuchta, Low-temperature mechanism of adsorption of S.M. Wang, J.H. Luo, A.W. Xu, Bifunctional pH-sensitive Zn(ii)-curcumin
methane: comparison between homogenous and heterogeneous pores, Col- nanoparticles/siRNA effectively inhibit growth of human bladder cancer cells
loids Surf 496 (2016) 86e93, https://doi.org/10.1016/j.colsurfa.2015.11.063. in vitro and in vivo, J. Mater. Chem. B 2 (2014) 2714e2724, https://doi.org/
[18] Stephen K. Brand, Yamil J. Colo  n, Rachel B. Getman, Randall Q. Snurr, Design 10.1039/c3tb21625j.
strategies for metal alkoxide functionalized metal-organic frameworks for [41] S. Wu, X. Ma, J. Ran, Y. Zhang, F. Qin, Y. Liu, Application of basic isoreticular
ambient temperature hydrogen storage, Microporous Mesoporous Mater 171 nanoporous metal-organic framework: IRMOF-3 as a suitable and efficient
(2013) 103e109, https://doi.org/10.1016/j.micromeso.2012.12.020. catalyst for the synthesis of chalcone, RSC Adv 5 (2015) 14221e14227,
[19] Marianna Kotzabasaki, Emmanuel Tylianakis, Emmanuel Klontzas, George https://doi.org/10.1039/C4RA16180G.
E. Froudakis, OH-functionalization strategy in Metal-Organic Frameworks for [42] X. Leng, X. Dong, W. Wang, N. Sai, C. Yang, L. You, H. Huang, X. Yin, J. Ni,
drug delivery, Chem. Phys. Lett. 685 (2017) 114e118, https://doi.org/10.1016/ Biocompatible Fe-based micropore metal-organic frameworks as sustained-
j.cplett.2017.07.053. release anticancer drug carriers, Molecules 23 (2018) 2490, https://doi.org/
[20] E.N. Koukaras, T. Montagnon, P. Trikalitis, Dimitrios Bikiaris, Aristides 10.3390/molecules23102490.
D. Zdetsis, George E. Froudakis, Toward efficient drug delivery through suit- [43] W. Liang, H. Xu, F. Carraro, N.K. Maddigan, Q. Li, S.G. Bell, D.M. Huang,
ably prepared metal-organic frameworks: a first-principles study, J. Phys. A. Tarzia, M.B. Solomon, H. Amenitsch, L. Vaccari, C.J. Sumby, P. Falcaro,
Chem. C 118 (2014) 8885e8890, https://doi.org/10.1021/jp410282m. C.J. Doonan, Enhanced activity of enzymes encapsulated in hydrophilic metal-
[21] S. Mura, J. Nicolas, P. Couvreur, Stimuli-responsive nanocarriers for drug de- organic frameworks, J. Am. Chem. Soc. 141 (2019) 2348e2355, https://doi.org/
livery, Nat. Mater. 12 (2013) 991e1003, https://doi.org/10.1038/nmat3776. 10.1021/jacs.8b10302.
[22] W.K. Kim, Y. Pyee, H.J. Chung, H.J. Park, J.Y. Hong, K.H. Son, S.K. Lee, Antitumor [44] W. Liang, P. Wied, F. Carraro, C.J. Sumby, B. Nidetzky, C.K. Tsung, P. Falcaro,
activity of spicatoside A by modulation of autophagy and apoptosis in human C.J. Doonan, Metal-organic framework-based enzyme biocomposites,
colorectal cancer cells, J. Nat. Prod. 79 (2016) 1097e1104, https://doi.org/ Chem. Rev. 121 (2021) 1077e1129, https://doi.org/10.1021/acs.chemrev.
10.1021/acs.jnatprod.6b00006. 0c01029.
[23] S. Li, C. Zhang, W. Cao, B. Ma, X. Ma, S. Jin, J. Zhang, P.C. Wang, F. Li, X.-J. Liang, [45] T. Xu, Y. Ma, Q. Yuan, H. Hu, X. Hu, Z. Qian, J.K. Rolle, Y. Gu, S. Li, Enhanced
Anchoring effects of surface chemistry on gold nanorods: modulating auto- ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform
phagy, J. Math. Chem. B 3 (2015) 3324e3330, https://doi.org/10.1039/ of ferrous hemoglobin for tumor synergistic therapy, ACS Nano 14 (2020)
C5TB00076A. 3414e3425, https://doi.org/10.1021/acsnano.9b09426.
[24] B. Joe, M. Vijaykumar, B. Lokesh, Biological properties of curcumin-cellular [46] G. Chen, J. Luo, M. Cai, L. Qin, Y. Wang, L. Gao, P. Huang, Y. Yu, Y. Ding, X. Dong,
and molecular mechanisms of action, Crit. Rev. Food Sci. Nutr. 44 (2004) X. Yin, J. Ni, Investigation of metal-organic framework-5 (MOF-5) as an anti-
97e111, https://doi.org/10.1080/10408690490424702. tumor drug oridonin sustained release carrier, Molecules 24 (2019) 3369,
[25] A. Giordano, G. Tommonaro, Curcumin and cancer, Nutrients 11 (2019) 2376, https://doi.org/10.3390/molecules24183369.
https://doi.org/10.3390/nu11102376. [47] Philip L. Ritger, A. Nikolaos, Peppas, A simple equation for description of so-
[26] A.S.S.R.A. Sharma, Curcumin: preventive and therapeutic properties in labo- lute release I. Fickian and non-fickian release from non-swellable devices in
ratory studies and clinical trials, Antioxid. Redox Signaling 10 (2008) the form of slabs, spheres, cylinders or discs, J. Contr. Release 5 (1987) 23e36,
511e545, https://doi.org/10.1089/ars.2007.1769. https://doi.org/10.1016/0168-3659(87)90034-4.
[27] T. Feng, Y. Wei, R.J. Lee, L. Zhao, Liposomal curcumin and its application in [48] R.A. Petros, J.M. Desimone, Strategies in the design of nanoparticles for
cancer, Int. J. Nanomed. 12 (2017) 6027e6044, https://doi.org/10.2147/ therapeutic applications, Nat. Rev. Drug Discov. 9 (2010) 615e627, https://
IJN.S132434. doi.org/10.1038/nrd2591.
[28] H. Liang, X. Sun, J. Gao, B. Zhou, Chitosan coordination driven self-assembly [49] E. Linnane, S. Haddad, F. Melle, Z. Mei, D. Fairen-Jimenez, The uptake of metal-
for effective delivery of curcumin, Int. J. Biol. Macromol. 165 (2020) organic frameworks: a journey into the cell, Chem. Soc. Rev. 51 (2022)
2267e2274, https://doi.org/10.1016/j.ijbiomac.2020.10.097. 6065e6086, https://doi.org/10.1039/d0cs01414a.
[29] Z. Li, Q. Lin, D.J. McClements, Y. Fu, H. Xie, T. Li, G. Chen, Curcumin-loaded [50] W.F. Liang, Y.X. Gong, H.F. Li, F.L. Sun, W.L. Li, D.Q. Chen, D.P. Xie, C.X. Ren,
core-shell biopolymer nanoparticles produced by the pH-driven method: X.Y. Guo, Z.Y. Wang, T. Kwon, H.N. Sun, Curcumin activates ROS signaling to
physicochemical and release properties, Food Chem 355 (2021), 129686, promote pyroptosis in hepatocellular carcinoma HepG2 cells, In Vivo 35
https://doi.org/10.1016/j.foodchem.2021.129686. (2021) 249e257, https://doi.org/10.21873/invivo.12253.
[30] L.M. Lim, K. Hadinoto, High-payload buccal delivery system of amorphous [51] J. Cao, L.P. Jiang, Y. Liu, G. Yang, X.F. Yao, L.F. Zhong, Curcumin-induced gen-
curcumin-chitosan nanoparticle complex in hydroxypropyl methylcellulose otoxicity and antigenotoxicity in HepG2 cells, Toxicon 49 (2007) 1219e1222,
and starch films, Int. J. Mol. Sci. 22 (2021) 9399, https://doi.org/10.3390/ https://doi.org/10.1016/j.toxicon.2007.02.006.
ijms22179399. [52] C. He, D. Liu, W. Lin, Nanomedicine applications of hybrid nanomaterials built
[31] N. Liu, Y. Lu, Y. Zhang, Y. Gao, L. Mao, Surfactant addition to modify the from metal-ligand coordination bonds: nanoscale metal-organic frameworks
structures of ethylcellulose oleogels for higher solubility and stability of and nanoscale coordination polymers, Chem. Rev. 115 (2015) 11079e11108,
curcumin, Int. J. Biol. Macromol. 165 (2020) 2286e2294, https://doi.org/ https://doi.org/10.1021/acs.chemrev.5b00125.
10.1016/j.ijbiomac.2020.10.115. [53] Jinyue Zeng, Xiaoshuang Wang, Xianzheng Zhang, Zhuo Renxi, Research
[32] L. Huang, H. Wang, J. Chen, Z. Wang, J. Sun, D. Zhao, Y. Yan, Synthesis, progress in functional metal-organic frameworks for tumor therapy, Huaxue
morphology control, and properties of porous metal-organic coordination Xuebao 77 (2019) 1156e1163, https://doi.org/10.6023/A19070259.
polymers, Microporous Mesoporous Mater 58 (2003) 105e114, https:// [54] R.S. Forgan, The surface chemistry of metal-organic frameworks and their
doi.org/10.1016/S1387-1811(02)00609-1. applications, Dalton Trans 48 (2019) 9037e9042, https://doi.org/10.1039/
[33] Hong Dong, Yang Gui-Xin, Xin Zhang, Xiang-Bin Meng, Sheng Jing-Li, Xiao- c9dt01710k.
Jun Sun, Feng Yu-Jie, Fengming Zhang, Folic acidfunctionalized Zr-based [55] T. Simon-Yarza, A. Mielcarek, P. Couvreur, C. Serre, Nanoparticles of metal-
metal-organic frameworks as drug carriers for active tumor-targeted, Chem. organic frameworks: on the road to in vivo efficacy in biomedicine, Adv.
Eur J. 24 (2018) 17148e17154, https://doi.org/10.1002/chem.201804153. Mater. 30 (2018), e1707365, https://doi.org/10.1002/adma.201707365.
[34] Mengru Cai, Wulin Liang, Kaixin Wang, Dongge Yin, Tingting Fu, Rongyue Zhu, [56] W. Xu, Y. Kang, L. Jiao, Y. Wu, H. Yan, J. Li, W. Gu, W. Song, C. Zhu, Tuning
Changhai Qu, Xiaoxv Dong, Jian Ni, Xingbin Yin, Aperture modulation of atomically dispersed Fe sites in metal-organic frameworks boosts peroxidase-
isoreticular metal organic frameworks for targeted antitumor drug delivery, like activity for sensitive biosensing, Nano-Micro Lett. 12 (2020) 184, https://
ACS Appl. Mater 14 (2022) 36366e36378, https://doi.org/10.1021/acsami. doi.org/10.1007/s40820-020-00520-3.
2c07450. [57] P. Horcajada, C. Serre, M. Vallet-Regí, M. Sebban, F. Taulelle, G. Fe rey, Metal-
[35] Sami Ullaha, Mohamad Azmi Bustamb, Mohammed Ali Assiria, G. Abdullah, organic frameworks as efficient materials for drug delivery, Angew Chem. Int.
A. Al-Sehemia Firas, Abdul Kareemc, Mukhtarb Ahmad, Muhammad Ayoubb, Ed. Engl. 45 (2006) 5974e5978, https://doi.org/10.1002/anie.200601878.
Girma Gonfad, Synthesis and characterization of iso-reticular metal-organic [58] M. AL Haydar, H.R. Abid, B. Sunderland, S. Wang, Metal-organic frameworks as
Framework-3 (IRMOF-3) for CO2/CH4 adsorption: impact of post-synthetic a drug delivery system for flurbiprofen, Drug Des. Dev. Ther. 11 (2017)
aminomethyl propanol (AMP) functionalization, J. Nat. Gas Sci. Eng. 72 2685e2695, https://doi.org/10.2147/DDDT.S145716.
(2019), 103014, https://doi.org/10.1016/j.jngse.2019.103014. [59] S. Boi, N. Rouatbi, E. Dellacasa, D. Di Lisa, P. Bianchini, O. Monticelli,
[36] Mengru Cai, Liuying Qin, Longtai You, Functionalization of MOF-5 with mono- L. Pastorino, Alginate microbeads with internal microvoids for the sustained
substituents: effects on drug delivery behavior, RSC Adv 10 (2020) release of drugs, Int. J. Biol. Macromol. 156 (2020) 454e461, https://doi.org/
36862e36872, https://doi.org/10.1039/d0ra06106a. 10.1016/j.ijbiomac.2020.04.083.
[37] David Dubbeldam Youn-Sang Bae, Andrew Nelson, Krista S. Walton, Joseph [60] Y.J. Meng, S.Y. Wang, Z.W. Guo, M.M. Cheng, J. Li, D.Q. Li, Design and prepa-
T. Hupp, Randall Q. Snurr, Strategies for characterization of large-pore metal- ration of quaternized pectin-Montmorillonite hybrid film for sustained drug
organic frameworks by combined experimental and computational methods, release, Int. J. Biol. Macromol. 154 (2020) 413e420, https://doi.org/10.1016/
Chem. Mater. 21 (2009) 4768e4777, https://doi.org/10.1021/cm803218f. j.ijbiomac.2020.03.140.
[38] G.a Chen, X.a Leng, J.a Luo, L.a You, C.a Qu, X.a Dong, H.c,d Huang, X.a Yin, [61] H. Zhang, W. Jiang, R. Liu, J. Zhang, D. Zhang, Z. Li, Y. Luan, Rational design of
J.a,b. Ni, In vitro toxicity study of a porous iron(III) metal-organic framework, metal-organic framework nanocarrier-based codelivery system of doxoru-
Molecules 24 (2019) 1211, https://doi.org/10.3390/molecules24071211. bicin hydrochloride/verapamil hydrochloride for overcoming multidrug
[39] Xue-Zhou Zhao, Teng Jiang, Long Wang, Hao Yang, Sui Zhang, Ping Zhou, resistance with efficient targeted cancer therapy, ACS Appl. Mater. Interfaces 9
Interaction of curcumin with Zn(II) and Cu(II) ions based on experiment and (2017) 19687e19697, https://doi.org/10.1021/acsami.7b05142.

12
M. Cai, B. Ni, X. Hu et al. Journal of Science: Advanced Materials and Devices 7 (2022) 100507

[62] D. Hu, H. Xu, B. Xiao, D. Li, Z. Zhou, X. Liu, J. Tang, Y. Shen, Albumin-stabilized photodynamic therapy, J. Am. Chem. Soc. 138 (2016) 3518e3525, https://
metal-organic nanoparticles for effective delivery of metal complex anti- doi.org/10.1021/jacs.6b00007.
cancer drugs, ACS Appl. Mater. Interfaces 10 (2018) 34974e34982, https:// [65] Dipranjan Laha, Kunal Pal, Angshuman Ray Chowdhuri, Pravat Kumar Parida,
doi.org/10.1021/acsami.8b12812. Sumanta Kumar Sahu, Kuladip Jana, Parimal Karmakar, Fabrication of
[63] M.D. Rowe, D.H. Thamm, S.L. Kraft, S.G. Boyes, Polymer-modified gado- curcumin-loaded folic acid-tagged metal-organic framework for triple-
linium metal-organic framework nanoparticles used as multifunctional negative breast cancer therapy in in vitro and in vivo systems, New J.
nanomedicines for the targeted imaging and treatment of cancer, Bio- Chem. 43 (2019) 217e229, https://doi.org/10.1039/C8NJ03350A.
macromolecules 10 (2009) 983e993, https://doi.org/10.1021/bm9 [66] S.Y. Li, B.R. Xie, H. Cheng, C.X. Li, M.K. Zhang, W.X. Qiu, W.L. Liu, X.S. Wang,
00043e. X.Z. Zhang, A biomimetic theranostic O2-meter for cancer-targeted photo-
[64] J. Park, Q. Jiang, D. Feng, L. Mao, H.C. Zhou, Size-controlled synthesis of por- dynamic therapy and phosphorescence imaging, Biomaterials 151 (2018)
phyrinic metal-organic framework and functionalization for targeted 1e12, https://doi.org/10.1016/j.biomaterials.2017.10.021.

13

You might also like