You are on page 1of 9

pubs.acs.

org/IC Article

Biocompatible MIL-101(Fe) as a Smart Carrier with High Loading


Potential and Sustained Release of Curcumin
Roya Karimi Alavijeh and Kamran Akhbari*
Cite This: https://dx.doi.org/10.1021/acs.inorgchem.9b02756 Read Online

ACCESS Metrics & More Article Recommendations *


sı Supporting Information
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via LA TROBE UNIV on February 25, 2020 at 03:23:18 (UTC).

ABSTRACT: The purpose of this study was the investigation of the potential of MIL-101(Fe) for load and sustained release of
curcumin (CCM), as an anticancer drug, with pH stimulus. The reasons for choosing this type of metal−organic framework (MOF)
are its high surface area, acceptable stability in a water medium, and its biocompatible components (iron and terephthalic acid) with
low toxicity to normal cells. The obtained results from UV−vis analysis confirmed that this MOF is a smart carrier with a higher
release rate in acidic pH (pH 5), which is a condition similar to that in cancer cells, than that at pH 7.4 (in normal cells). Therefore,
this MOF is a pH-stimulus-controlled release carrier with 56.3% drug loading content and sustained drug release over 22 days. In
order to evaluate the cell viability after treatment with free CCM, MIL-101(Fe), and MIL-101(Fe)@CCM, the cytotoxicity
investigation using MTT assays was performed against HeLa and HEK 293 cell lines up to 48 h. Obtained results showed that MIL-
101(Fe)@CCM exhibited more cell growth inhibition effect on HeLa cells in comparison with HEK 293. One of the reasons for the
high loading and sustained release of CCM was surface adsorption of this drug and its interactions with open metal sites in MIL-
101(Fe). In the end, the kinetic models of drug release were evaluated, and the obtained results showed that in this case diffusion is
the main driving force for the drug release process.

1. INTRODUCTION their high stability. Therefore, it is difficult to degrade and


One of the leading causes of death in the world is cancer. The subsequently remove after drug release process.8 Metal−
main problems for overcoming this issue are poor delivery of organic frameworks constructed from organic linkers and
cancer therapeutics and their inadequate distribution in tumor inorganic nodes with features such as ultrahigh porosity, high
cells. A promising strategy to address this challenge is the use surface area, and thermal stability are appropriate candidates in
of nanocarriers because they can increase drug circulation many fields such as catalyst,9 gas storage,10 synthesis of
times and improve the safety profile.1 In recent years, nanomaterials,11 chemical sensors12 antibacterial activity,13 and
pharmaceutical industries are focusing on the application of especially drug delivery.14 Among reported MOFs, iron
sustained-release formulations that include any drug delivery carboxylate MOFs are good candidates for biological
system with the ability to slow release of drug molecules over application because of their low toxicity and biodegradability.
an extended period. These systems lead to reduced side MIL-101(Fe) is a mesoporous carrier with pore sizes of 12 Å ×
effects.2 There are many organic and inorganic compounds 29 Å; 16 Å × 34 Å and composed of nontoxic components.15
which are candidates for use as drug delivery systems (DDS) This MOF is constructed from terephthalic acid (H2BDC) as
such as nanoemulsions,3 hydrogels,4 micelles,5 liposomes,6 and
mesoporous silica.7 These candidates have both advantages Received: September 16, 2019
and some drawbacks. For example, organic carriers are
biocompatible, but in some cases their loading capacities are
low. Inorganic carriers with well-defined porosity have high
loading capacity, but the main problem of these materials is

© XXXX American Chemical Society https://dx.doi.org/10.1021/acs.inorgchem.9b02756


A Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

an organic linker and iron as an inorganic node. Iron exists in 208S). Fluorescence images were obtained by OPTIKA B500TiFL
blood plasma at approximately 22 μM. According to kinds of microscope.
literature, both components show little toxicity in the body. 2.2. Synthesis of MIL-101(Fe). MIL-101(Fe) was synthesized
according to previous literature.18 Briefly, 2 mmol of FeCl3.6H2O, 2
Therefore, based on the above mentioned reasons, we have
mmol of terephthalic acid and 1.8 mL of acetic acid were mixed in 50
chosen MIL-101(Fe) as a carrier for delivery of curcumin as an mL of N,N-dimethylformamide (DMF). This mixture was refluxed at
anticancer drug. Curcumin (CCM) is a natural chemo- 110 °C for 24 h. At the end of the reaction, the brick-red precipitated
therapeutic agent which is a pigment from turmeric. This product was isolated with the centrifuge. The yield of the reaction was
compound has attracted considerable attention because of its 78% (based on final product). Then the obtained powders were
therapeutic effects such as antitumor, antioxidant, and washed with DMF and stored in this solvent for 2 days (the solvent
antibacterial activities. The diketo group in the curcumin was refreshed two times each day). This process was repeated with
structure exhibits keto−enol tautomerism (Scheme S1). CCM methanol instead of DMF. In the end, the product was heated at 80
°C for 1 day for activation.
is hydrophobic molecule, insoluble in water but soluble in 2.3. Drug Loading. First, 50 mg of activated MIL-101(Fe) was
polar solvents such as DMSO, ethanol, methanol, and suspended in 50 mL of a concentrated solution of CCM (2.65
chloroform. Aqueous curcumin solution is achievable by mg.mL−1) in ethanol and stirred for 48 h in the dark. After 48 h, the
adding surfactants, lipids, cyclodextrins, albumins, biopoly- solution was centrifuged, and the obtained products were washed with
mers, and so on, although using surfactant is the best method ethanol three times. The amount of drug loading was obtained based
for preparing high concentration solutions of curcumin in on the reported method in previous literature.16a,19 The obtained
water. One of the features of curcumin is its chemical reactivity MIL-101(Fe)@CCM was separated and dried in the air at room
with reactive oxygen species (ROS) that consists of free radical temperature. To determine the amount of drug loading in the MOF, 3
mg of MIL-101(Fe)@CCM was digested in 0.75 mL of HCl (2 M)
oxidants and molecular oxidants. There are active sites in the and diluted to 30 mL with ethanol. Measurements were done by UV−
structure of curcumin with the ability to reduce ROS, and the vis spectrophotometer based on the absorbance of the solution at 425
results obtained from investigations by different groups nm. A calibration curve of CCM in ethanol was used for obtaining
revealed that phenolic O−H which is more efficient in the DLC (drug loading content) and DLE (drug loading efficiency)
reduction of ROS leads to the formation of phenoxyl radicals (Figure S1).
that are less reactive species than earlier ROS. This phenoxyl 2.4. In Vitro Drug-Release Studies. First, 2 mg of MIL-101(Fe)
radical can convert to curcumin by water-soluble antioxidants @CCM (dry sample) was suspended in 40 mL of phosphate-buffered
such as ascorbic acid. Therefore, this compound has solution (PBS) + 1% v/v Tween 80 as media and placed in the shaker
(with speed of 80 rpm) to determine drug release profile. Tween 80
antioxidant and anticancer activities, but the main problem was used as stabilizer and solubilizer of CCM because CCM is not
for its administration is due to hydrophobicity and poor stable in PBS. At a predetermined time, 1 mL of the solution was
bioavailability. Therefore, various efforts have been made to removed and replaced with 1 mL of fresh preheated PBS + 1% v/v
encapsulate CCM.16 There are many studies on the develop- Tween 80 to maintain constant conditions. The withdrawn solution
ment of controlled DDS. Some of them consist of stimuli such was analyzed with UV−vis spectrophotometer to obtain the
as pH, light irradiation, magnetic field, and temperature concentration of released drug based on the calibration curve of
increasing. Among them, pH variation is the most widely used CCM in PBS+ 1% v/v Tween 80. The obtained results were reported
as an average from two repeated experiments. Then, the percent
stimulus to induce the release of active molecules in a medium
accumulative drug release was calculated as a function of time.
with a controlled manner. In this case, two factors are effective: Equations for the calculation of corrected drug concentration20 are
(I) the solubility of the active molecule and (II) the stability of listed in the Supporting Information. These calculations were done at
the carrier.17 In this work, the drug loading capacity of MIL- two pHs (7.4 and 5).
101(Fe) for curcumin as an anticancer drug considering pH 2.5. Cytotoxicity Assay. The toxicity of MIL-101(Fe), CCM, and
stimulus condition was investigated (pH 7.4 for normal tissue MIL-101(Fe)@CCM on HeLa and HEK 293 cell lines was
and pH 5 for simulation of cancer cells condition). In the end, investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium-
kinetic analysis of drug release was studied. On the basis of the bromide (MTT) assay. Briefly, the HeLa and HEK 293 cells with a
density of 104 cells/well were seeded in 96-well plates and incubated
results obtained at the two pH values, it can be concluded that overnight. Various concentrations of MIL-101(Fe)@CCM, CCM,
MIL-101(Fe) is a pH-sensitive carrier and can act as a smart and MIL-101(Fe) in DMEM were prepared and supplemented with
DDS with a sustained-release manner. 10% FBS. These solutions were added to 5 wells containing the
seeded cells. In the following 24 h, the medium was exchanged with
2. MATERIALS AND METHODS 100 μL of a 0.5 mg mL−1 MTT solution (Sigma, USA). After 4 h,
formazan crystals were dissolved in 100 μL of isopropanol (Merck,
2.1. Materials and Techniques. All chemicals were used as Germany) by incubation at 37 °C for 15 min. The absorbance of each
commercially obtained and without further purification. IR spectra well was evaluated by a multiwell microplate reader (Statfax 2100,
were recorded using an Equinox 55 FT-IR spectrometer (Bruker, USA) at 545 nm then normalized to the control test.
Bremen, Germany) in ATR form, in the range of 400−4000 cm−1 2.6. Cellular Uptake Studies. Because of the intrinsic green
with 4.0 cm−1 resolution and 16 scan numbers. UV−vis spectra were fluorescence of CCM, the cellular uptake of MIL-101(Fe)@CCM was
recorded by Rayleigh spectrophotometer in the range of 300−550 investigated by fluorescence microscopy. For this analysis, HeLa cells,
nm. The PXRD patterns were recorded with a Rigaku Ultima IV with a density of 104 cells per well, were seeded in 6-well culture
powder diffractometer using Cu Kα radiation (λ = 1.54056 Å) with plates and incubated in DMEM for 24 h. After this time, the culture
step-size of 0.01671°. Simulated XRD powder patterns based on medium was replaced by a solution of CCM in fresh DMEM with a
single-crystal data were prepared using the Mercury software. BET concentration of 20 μg/mL. After 48 h, the medium was removed and
surface area MIL-101(Fe) and MIL-101(Fe)@CCM were calculated cells washed with PBS (pH 7.4). The cells were fixed with 800 μL of
from the N2 adsorption isotherms at 77K with Belsorp mini II 4% formaldehyde and washed with PBS again. Then samples were
apparatus. ZP was measured with Zetasizer Nano Series, Malbern measured by fluorescence microscopy.
ZS90 Instrument at water medium. The samples were characterized 2.7. Kinetic Study. Mathematical models are useful tools to study
with a scanning electron microscope (Philips XL 30) with gold the optimal design of new pharmaceutical formulations, evaluate drug
coating and transmission electron microscopy (TEM Philips EM release processes, and understand drug release mechanism.21 Some

B https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

main-release kinetic models were applied to the data obtained in the


drug release studies to predict the release process. The zero-order,
first-order, Higuchi, Hixson−Crowell, and Korsmeyer−Peppas are
well-known mathematical models that were used in many kinds of
literature.22 In this study, kinetic models were investigated to achieve
a better understanding of the drug release mechanism. These models
with their formula are available in Table S1.

3. RESULTS AND DISCUSSIONS


3.1. Synthesis and Characterization of MIL-101and
MIL-101(Fe)@CCM. MIL-101(Fe) was prepared in DMF
solution at a molar ratio of Fe3+/BDC2− = 1:1 under reflux
condition. MIL-101(Fe), activated MOF, and MIL-101(Fe)@
CCM are represented in Figure 1.

Figure 2. PXRD of simulated pattern of MIL-101(Cr), activated MIL-


101(Fe), and MIL-101(Fe)@CCM.

Figure 1. Photograph of MIL-101(Fe) before and after activation and


MIL-101(Fe)@CCM.

The FT-IR spectra of MIL-101(Fe), activated MIL-101(Fe),


MIL-101@CCM, and pure curcumin are shown in Figure S2.
A peak at 1657 cm−1 (represented with a red rectangle) is
attributed to a CO group of DMF molecule that is omitted
after activation with methanol exchange and heating in the
oven.23 There are some characteristic peaks in curcumin
spectra that also appeared in MIL-101(Fe)@CCM that
represent the existence of CCM in the structure. These
peaks are listed in Table S2 and shown in Figure S2 with green
dots.
Other evidence of CCM presence in MIL-101(Fe) is 1H
NMR (Figure S3). There are some characteristic peaks in the
spectrum of MIL-101(Fe)@CCM in the region of 6.5−7.5
ppm and at 3.78 ppm that are attributed to CCM24 (peak at
2.5 ppm is assigned to DMSO-d6). The presence of these peaks Figure 3. Nitrogen adsorption−desorption isotherm of MIL-101(Fe)
indicates that curcumin was present in MIL-101(Fe)@CCM. before and after CCM loading.
Powder X-ray diffraction (PXRD) patterns of activated MIL-
101(Fe) and MIL-101(Fe)@CCM are shown in Figure 2.
Both patterns are matched to the simulated PXRD pattern of
MIL-101(Cr). This result confirmed the successful synthesis,
activation, and drug loading of MIL-101(Fe), and this pattern
suggests that activation of MIL-101(Fe) and CCM incorpo-
ration did not affect the MIL-101(Fe) crystal structure.
The results obtained from nitrogen sorption measurements
showed reduced the BET surface area of MIL-101(Fe) after
the drug loading process. The BET surface area of MIL-
101(Fe) after activation was 1419.9 m2/g, but after drug
loading, this amount was reduced to 84.09 m2/g. Moreover,
the total pore volume decreased from 1.06 to 0.64 cm3 g−1).
This reduced surface area and changed N2 isotherm (Figure 3)
represents CCM loading in the pores of MIL-101(Fe).
The ZP (zeta potential), also known as electrokinetic
potential, is defined as the amount of potential at the slipping/
shear plane of moving colloidal particles under an electric field.
The electric potential of the surface of particles is defined as
“the amount of work that needs to be done to bring a unit Figure 4. Zeta potential of MIL-101(Fe) before and after CCM
loading.
positive charge from infinity to the surface without any
acceleration.”25 ZP measurements in Figure 4 indicated a
positive zeta potential of +8.3 mV for MIL-101(Fe)@CCM,
C https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

Figure 5. SEM images of (A) MIL-101(Fe) before, (B) after CCM loading, and (C) TEM image of MIL-101(Fe)@CCM.

which is lower than MIL-101 (+28.6 mV). On the basis of the According to previous literature,19a,27 curcumin could
previously reported literature,26 this reason can be attributed to coordinate with the central metal in the diketo form. In this
the interaction of the electrons in the β-diketones of CCM study, some evidence confirms that there are some interactions
with metal ions in the external surface of MOF. Therefore, the between the open metal sites in MIL-101(Fe) and CCM. The
zeta potential of MIL-101(Fe) is decreased after the SBU of MIL-101(Fe) is composed of a carboxylate-bridged,
incorporation of CCM to form MIL-101(Fe) @CCM. This oxo-centered, trinuclear Fe3+ complex (Figure 6). This SBU
result represents that beside encapsulation of drug into the
pores of the MIL-101 (Fe) that leads to a decrease in surface
area and pore volume of the MOF the surface adsorption of
curcumin has also been effective in drug loading content.
The SEM images before and after drug loading are shown in
Figure 5a,b. SEM image of MIL-101(Fe)@CCM (Figure 5b)
with agglomerated nanostructures is different from MIL-
101(Fe) with a nanostructured surface. A comparison between
these images also approved the surface adsorption of CCM in
addition to its internal loading. Also, the TEM image of MIL-
101(Fe) after drug loading (Figure 5c) clearly supported the Figure 6. SBU of MIL-101(Fe).
nanosize of MIL-101(Fe)@CCM and surface adsorption of
CCM. consists of removable coordinated H2O molecules. After
3.2. Drug Loading and Release Study. The amount of activation, coordinative unsaturated sites (CUSs) or open-
drug loading was obtained based on the calibration curve of metal sites are formed.28 These CUSs are capable to
CCM in ethanol with UV−vis spectrophotometer in λ = 425 coordinate to CCM.
nm that is due to π−π* transitions (eqs 1 and 2 in the The FT-IR spectrum in Figure S2 shows two bands at 1496
Supporting Information). According to calculations, DLC and and 1424 cm−1 that indicate the presence both of the free
DLE are 56.3 and 48.7%, respectively. carbonyl group of the diketone and enol form in CCM.
D https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

Figure 7. Cumulative drug release profile in two pH of 7.4 and 5.

Scheme 1. Proposed Mechanism for Higher Release of CCM at Acidic pH

Figure 8. (A) In vitro biocompatibility of MIL-101(Fe) (with concentrations of 400, 200, 100, 50, 25, and 12.5 μg/mL), CCM with concentrations
of: 250, 125, 62, 31, 16, and 8 μg/mL), and MIL-101(Fe)@CCM (with concentrations of: 444, 222, 111, 55.5, 27.75, and 13.875 μg/mL) against
HeLa cell lines for 48 h. Selectin of concentrations was based on the amount of drug loading on MIL-101(Fe). (B) In vitro biocompatibility of
MIL-101(Fe)@CCM against HeLa and HEK cell lines at various concentrations for 48 h.

However, after the drug loading step, the peak at the lower of the MIL-101(Fe). These drugs probably have some
wavelength (1424 cm−1, shown with a red circle in Figure S2) interactions with metal ions27,29 that lead to the slow release
that is attributed to the enol form has been omitted. Also, the of CCM and high drug loading content.
broad peak (that contributed to enolic OH, shown with a red Another reason to support the coordination between metal
circle in Figure S2) in the range of 3200−3500 cm−1 is absent and CCM is 1H NMR analysis is reported previously by Tiwari
after drug loading. Therefore, it concluded that CCM in MIL- and co-workers.19a The 1H NMR spectrum of curcumin
101(Fe)@CCM is in the diketo form. On the basis of ZP displayed peaks at δ 6.06 ppm in DMSO + DCl solutions
results that showed reduced potential after CCM loading, it is (Figure S4) that are related to methine protons and confirmed
concluded that beside drugs loaded in the pores of MIL- the presence of enol form of curcumin. This peak was absent in
101(Fe) that was confirmed with reduced BET surface area the MIL-101(Fe)@CCM spectrum. It could be related to the
and pore volume, there are some drugs adsorbed on the surface interactions between Fe and the diketo form of curcumin.
E https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

Table 1. Results Obtained from Investigation of Different Models in Terms of R2


pH zero-order first-order Higuchi model Korsmeyer−Peppas Hixson−Crowell
7.4 R2 = 0.864 R2 = 0.9019 R2= 0.9783 R2 = 0.9729 n = 0.28 R2 = 0.921
5 R2= 0.8393 R2 = 0.9364 2
R = 0.9716 R2 = 0.9957 n = 0.41 R2 = 0.9094

In the following, the in vitro drug release profile of MIL- the initial amounts of the drug, the amount of drug released at
101(Fe)@CCM was investigated (based on the calibration time t, and the zero-order rate constant.
curve of CCM in PBS + 1% v/v Tween 80 and eqs 3−5 in the 3.4.2. First-Order Model. The first-order model represents
Supporting Information) at pHs of 7.4 and 5, and its results are that the drug release from a formulation depends upon two
shown in Figure 7. As shown in this picture, MIL-101(Fe)@ factors; I) time and II) initial concentration of the drug.
CCM presented a prolonged drug release profile, only 26 ± 2% Because of the dependence of the amount of drug released to
of CCM, even after 22 days in normal PBS (pH 7.4). This the amount of remaining drug in the matrix, the amount of
amount was higher in acidic pH, and the amount of cumulative active released tends to decrease in function of time. Log of the
drug release was reached to 64.7 ± 2.9% at that time. Also, the cumulative remaining drug (%) against time was used for
photograph in the inset of Figure 7 clearly shows the color fitting data obtained into this model. k1 in the formula in Table
difference at the two pHs. This slow release can prove the S1 represents the first-order rate constant.
attractive interaction between CCM in the diketo form with 3.4.3. Higuchi Model. This model is based on some
the metal ion of MIL-101(Fe). hypotheses: (I) The initial drug concentration in the matrix is
This pH-stimulus release feature of MIL-101(Fe) can reduce much higher than the solubility of the drug. (II) The diffusion
premature drug release in normal cells and enhance drug is unidirectional. In other words, drug diffusion takes place
release in the acidic microenvironments of tumors cells. This only in one dimension, and the edge effect is negligible. (III)
feature will be beneficial for cancer treatment. The drug particles are smaller than the system thickness. (IV)
The observed higher release of curcumin in acidic pH might The matrix swelling or dissolution are negligible. (V) The drug
be attributed to the protonation of CCM at lower pH.30 CCM diffusivity is constant. (VI) The sink conditions are attained in
can react with excess protons in solution. It is proved in the release environment. The Higuchi model indicates that the
published literature in 2014 that the diketo oxygen atoms can rate of drug release is based on the diffusion process according
act as a proton sponge. It means that the added protons lead to to Fick’s law. This model represents that the concentration of
a stable six-membered ring formation.31 Therefore, acidic pH active agent is proportional to the square root of time. KH in
can trigger the release of CCM. The proposed mechanism for this model is the release constant of Higuchi.
the higher release of CCM in the acidic condition is 3.4.4. Hixson−Crowell Model. Hixson and Crowell in 1931
represented in Scheme 1. discovered that a group of particles’ regular area is proportional
3.3. In Vitro Cytotoxicity by the MTT. The cytotoxicity to the cube root of its volume. On the basis of this finding, they
of MIL-101(Fe)@CCM, CCM, and MIL-101(Fe) were proposed an equation that is listed in Table S1. The drug
investigated by carrying out MTT tests on HeLa and HEK release from particles in this model is described keeping in
293 cell line. The obtained results (with seven repetitions) are view the volume and area of the particle. In the Hixson−
shown in Figure 8A,B. Based on the obtained results in Figure Crowell formula, Mt/M∞, and kHC are the fractional drug
8A, MIL-101(Fe) has no apparent cytotoxicity in the HeLa cell release and Hixson−Crowell rate constant, respectively. In the
line even up to high concentrations (400 μg/mL). However, Hixson−Crowell model, the dissolution velocity and not
MIL-101(Fe)@CCM exhibited a cell growth inhibition effect diffusion limit the drug release, which can occur through the
on HeLa cells with an IC50 72.6 μg/mL. A noticeable point in polymeric matrix. Data from the release study of the drug were
this figure is the greater effect of free CCM than MIL-101(Fe) fitted into Hixson−Crowell model by plotting cube root of
@CCM. This result is predictable. This in vitro analysis was percent of drug remaining against time.
done after 48 h, but MIL-101(Fe) needs 22 days to release its 3.4.5. Korsmeyer−Peppas Model. This model could be
entire drug load. In another word, based on the data obtained used to predict the drug release mechanism from a polymeric
in Figure 8, at 48 h only 27 and 10% of all drug load can be system. This equation was applied to the data by plotting the
released at pH 5 and 7.4, respectively. An important point in log of the cumulative drug release (%) against the log of time.
DDS is the amount of cytotoxicity to normal cells. Therefore, Mt/M∞, kKP, and n represent the fractional drug release, the
MIL-101(Fe)@CCM was tested on HeLa (as cancer cell Korsmeyer−Peppas rate constant, and release exponent,
model) and HEK 293 (as normal cell model) and the results respectively. The size of n characterizes the mechanism of
are shown in Figure 8B. As this picture indicates, the MIL- the drug release (Table S3). To find out the size of n, Mt/M∞
101(Fe)@CCM has no cytotoxicity to HEK 293 in low < 0.6 should only be used.
concentration. Even at high concentrations, its cytotoxicity is In the Fickian model (n = 0.5), the drug release is governed
very small in comparison with that in the HeLa cell line. by diffusion. A value of n = 1 represents the model is non-
3.4. Kinetic Models. The results obtained from the drug Fickian, and the drug release rate corresponds to zero-order
release profiles were fitted to some well-known kinetic models release kinetics. In this case, the swelling or relaxation of
that are briefly explained below. polymeric chains is the driving force for the drug release
3.4.1. Zero-Order Model. The zero-order kinetics model process. When 0.5 < n < 1, the model is non-Fickian or
represents that the release of an active agent is only a function anomalous transport. In this case, the drug release mechanism
of time with a constant rate and is independent of the time or is governed by diffusion and swelling. Finally, the Super Case II
concentration. The data obtained from drug release inves- model is characterized by n > 1, constituting an extreme form
tigation were fitted into this model by plotting a cumulative of drug transport. Tension and breaking of the polymer
drug release (%) against time. In this model M0, Mt and k are occur.21,22,22c,32
F https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

The results obtained from the investigation of the in vitro Table 2. List of Reported MOFs for Load and Release of
drug release profile with different mathematical models CCM
(Figures S5 and S6) are listed in Table 1. In acidic pH, the
MIL CCM loading time stimulus ref
correlation coefficient (R2) is higher for the Korsmeyer−
Peppas model, and at normal pH, this value is approximately The DLC and DLE were
ZIF-8 12.7% and 88.2%, respec- 100 (h) pH 16a
the same for the Higushi and Korsmeyer−Peppas models and tively.
slightly higher in the Higuchi model. As mentioned above, the The DLC and DLE were
MOF- pH and
Higuchi model indicates that the rate of drug release is based 11.8 and 78.7%, respec- 24 (h) 26
Zr(DTBA) GSHa
tively
on the diffusion process, according to Fick’s law. Moreover, the
The drug loading capacity
mount of n in acidic pH represents the quasi-Fickian diffusion ZIF-8
was 3.42%.
72 (h) pH 19a
mechanism. According to these results, it could be suggested The bulk loading of CCM
Ca-BDC 350 (min) pH 27
that the diffusion is main driving force for the drug release was about 0.14 g·g−1.
from MIL-101(Fe) at both pHs. PDA-MSN@ZIF
The loading capacity was
8/DOX + ∼32 (h) pH 34
The stability of the MIL-101(Fe)@CCM in PBS at pH 5 778 μg mg−1.
CUR
was investigated in PBS + 1% v/v Tween 80 for 7 days at 37 The DLC and DLE were
°C. After this time, the PXRD analysis was done (Figure S7). MCH NPsb 33.0% and 94.3%, respec- ∼72 h pH 29
On the basis of the obtained results, most of the main peaks tively.
are unchanged (represented with green color). These results CCM@ZIF-8/ The capacity of the encap-
∼180 h pH 19b
HAc sulated CCM was 4.24%.
indicate the stability of MIL-101(Fe) in the PBS solution after Fe3O4@Bio-
a week and confirm the proposed mechanism of drug release CCM was the organic linker. ∼50 h pH 35
MOF
obtained from kinetic study. However, based on the reported MIL-101(Fe)@ DLC and DLE are 56.3 and
528 (h) pH
this
literatures, beside the importance of the stability of drug CCM 48.7%, respectively. work
a
carriers in medical applications, a carrier with a degree of Glutathione. bCoating MC NPs (curcumin-loaded MIL-100) with
instability leads to avoiding accumulation of endogenous. For HA-PDA (PDA-modified). cHyaluronic acid (HA)-coated ZIF-8
example, it is expected that a carrier of anticancer drug, save its nanocomposite.
structure before reache to the tumor site. On the basis of
earlier reports, MIL-101(Cr) degraded after 7 days (in coordination of CCM to open metal sites on MIL-101(Fe)
simulated body fluid (SBF) at 37 °C).33 The stability of external surface. Coordinated CCM on the surface of MIL-
MIL-101(Fe)@CCM in PBS/Tween 80 at pH 5 at 37 °C was 101(Fe) presents the slow release of CCM from the pores of
investigated, and the PXRD pattern is shown in Figure S7. MIL-101(Fe). At acidic pH, because of the proposed
This pattern shows acceptable stability after a week. After mechanism (Scheme 1), the release of CCM adsorbed on
this time the MIL-101(Fe)@CCM starts to lose its crystal the external surface of MIL-101(Fe) was faster than that at
structure. Therefore, based on the results obtained in this study normal pH. Then the stored CCM in the pores of MIL-
it could be concluded that MIL-101(Fe)@CCM keeps its 101(Fe) releases more easily than at normal pH.
structure intact and that diffusion is a main driving force for On the basis of this comparison, it is clear that MIL-101(Fe)
the drug release process. MIL-101(Fe) is able to control has an appropriate potential for load and release of CCM. The
release for 22 days. Finally, after this long time, based on the reported data revealed the ability of this MOF for achieving
PXRD patterns, MIL-101(Fe) starts to lose the crystal suitable DDS with a pH-stimulus-controlled release manner.
structure that is suitable for avoiding endogenous accumulation
in the body. The SEM image of MIL-101(Fe)@CCM after 4. CONCLUSION
drug release (22 days) at pH 7.4 and 5 are shown in Figure S8 In this study, MIL-101 (Fe) was used for load and sustained
shows the nanosized structure with similar morphology to release of CCM as an anticancer drug. Obtained data from
fresh MIL-101(Fe)@CCM. drug release investigation in two pH (7.4 and 5) were fitted
The results obtained from the investigation of cellular with some mathematical models to understand the drug release
uptake of MIL-101(Fe)@CCM by fluorescence microscopy mechanism. This kind of MOF was chosen because of the low
are shown in Figure 9. Green fluorescence is detected which toxicity of its components, high surface area, large pore size,
indicates the endocytosis ability of MIL 101(Fe)@CCM that is and simple preparation. The results confirmed the ability of
related to its nanosized structure. MIL-101(Fe) for high loading of CCM (56.3 wt %) as an
There are some reports about the application of MOFs for anticancer drug with pH stimulus release behavior (more drug
load and release of CCM. In Table 2, MIL-101(Fe) is release in lower pH) over prolong time (22 days). On the basis
compared with other kinds of literature in the viewpoint of two of observed changes in IR, SEM, TEM, 1H NMR, and ZP
factors: drug release time and drug loading ability. It seems results before and after the drug loading process, the reasons
that the effective factor for its excellent CCM release time is for high loading and prolonged drug release time in
comparison with other reports can be attributed to the large
pore size of MIL-101(Fe) and combination of encapsulation of
drug molecules in the pores of MIL-101(Fe) and surface
adsorption of CCM on the open metal sites of this MOF. The
mathematical models showed that drug release data at pH 7.4
and 5 are best fitted by Higuchi (R2 = 0.9783) and
Korsmeyer−Peppas (R2 = 0.9957) models, respectively.
Figure 9. Fluorescence images of (A) MIL-101(Fe)@CCM (green These results imply that diffusion is main driving force for
fluorescence), (B) cell nucleus (blue fluorescence, stained by Hoechst CCM release from MIL-101(Fe)@CCM. MTT test on HEK
33342), and (C) the overlay image (scale bars = 10 μm). 293 cell line, as healthy cells model, indicates low cytotoxicity
G https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

of MIL-101(Fe)@CCM. A combination of high drug loading (5) (a) Gothwal, A.; Khan, I.; Kesharwani, P.; Chourasia, M. K.;
content, sustained-release behavior, and sensitivity to pH Gupta, U., Micelle-Based Drug Delivery for Brain Tumors. In
conditions (higher drug release at a pH similar to cancer cells) Nanotechnology-Based Targeted Drug Delivery Systems for Brain
can provide new opportunities to achieve more effective Tumors; Elsevier: 2018; pp 307−326. (b) Wang, Z.; Deng, X.;
Ding, J.; Zhou, W.; Zheng, X.; Tang, G. Mechanisms of drug release
treatment for cancer in the future.


in pH-sensitive micelles for tumour targeted drug delivery system: A
review. Int. J. Pharm. 2018, 535, 253−260. (c) Yi, Y.; Lin, G.; Chen,
ASSOCIATED CONTENT S.; Liu, J.; Zhang, H.; Mi, P. Polyester micelles for drug delivery and
*
sı Supporting Information cancer theranostics: current achievements, progresses and future
The Supporting Information is available free of charge at perspectives. Mater. Sci. Eng., C 2018, 83, 218−232.
https://pubs.acs.org/doi/10.1021/acs.inorgchem.9b02756. (6) (a) Franzé, S.; Marengo, A.; Stella, B.; Minghetti, P.; Arpicco, S.;
Cilurzo, F. Hyaluronan-decorated liposomes as drug delivery systems
Structure of curcumin, calibration curve, Table of IR for cutaneous administration. Int. J. Pharm. 2018, 535, 333−339.
peaks, kinetic models and their mathematical formula, (b) Shen, S.; Huang, D.; Cao, J.; Chen, Y.; Zhang, X.; Guo, S.; Ma,
1
H NMR spectra, PXRD patterns, calculations of drug W.; Qi, X.; Ge, Y.; Wu, L. Magnetic liposomes for light sensitive drug
loadin and release, curves of the application of drug delivery and combined photothermal-chemotherapy of tumor. J.
release data on mathematical models, and SEM images Mater. Chem. B 2019, 7, 1096−1106. (c) Xiao, W.; Fu, Q.; Zhao, Y.;
(PDF) Zhang, L.; Yue, Q.; Hai, L.; Guo, L.; Wu, Y. Ascorbic acid-modified
brain-specific liposomes drug delivery system with “lock-in” function.

■ AUTHOR INFORMATION
Corresponding Author
Chem. Phys. Lipids 2019, 224, 104727.
(7) (a) Fan, N.; Liu, R.; Ma, P.; Wang, X.; Li, C.; Li, J. The On-Off
chiral mesoporous silica nanoparticles for delivering achiral drug in
Kamran Akhbari − School of Chemistry, College of Science, chiral environment. Colloids Surf., B 2019, 176, 122−129. (b) Gao, Z.;
Shi, T.; Li, Y.; You, C.; Cheng, K.; Sun, B. Mesoporous silica-coated
University of Tehran, Tehran, Iran; orcid.org/0000-0002- gold nanoframes as drug delivery system for remotely controllable
4574-683X; Phone: +98 21 61113734; Email: akhbari.k@ chemo-photothermal combination therapy. Colloids Surf., B 2019,
khayam.ut.ac.ir; Fax: +98 21 66495291 176, 230−238. (c) Vallet-Regí, M.; Colilla, M.; Izquierdo-Barba, I.;
Manzano, M. Mesoporous silica nanoparticles for drug delivery:
Author current insights. Molecules 2018, 23, 47.
Roya Karimi Alavijeh − School of Chemistry, College of Science, (8) Orellana-Tavra, C.; Marshall, R. J.; Baxter, E. F.; Lázaro, I. A.;
University of Tehran, Tehran, Iran Tao, A.; Cheetham, A. K.; Forgan, R. S.; Fairen-Jimenez, D. Drug
Complete contact information is available at: delivery and controlled release from biocompatible metal−organic
https://pubs.acs.org/10.1021/acs.inorgchem.9b02756 frameworks using mechanical amorphization. J. Mater. Chem. B 2016,
4, 7697−7707.
(9) (a) Li, Y.; Zhang, X.; Xu, P.; Jiang, Z.; Sun, J. The design of a
Notes
novel and resistant Zn (PZDC)(ATZ) MOF catalyst for the chemical
The authors declare no competing financial interest.


fixation of CO2 under solvent-free conditions. Inorg. Chem. Front.
2019, 6, 317−325. (b) Rojas-Buzo, S.; García-García, P.; Corma, A.
ACKNOWLEDGMENTS Zr-MOF-808@ MCM-41 catalyzed phosgene-free synthesis of
Support of this investigation by the Iran National Science polyurethane precursors. Catal. Sci. Technol. 2019, 9, 146−156.
Foundation (INSF) under grant number 98002623 is (c) Noori, Y.; Akhbari, K. Post-synthetic ion-exchange process in
gratefully acknowledged. The authors also would like to nanoporous metal−organic frameworks; an effective way for
modulating their structures and properties. RSC Adv. 2017, 7,
acknowledge the financial support of the University of Tehran
1782−1808.
for this research under grant number 01/1/389845 for (10) (a) Li, H.; Wang, K.; Sun, Y.; Lollar, C. T.; Li, J.; Zhou, H.-C.
supporting this investigation.


Recent advances in gas storage and separation using metal−organic
frameworks. Mater. Today 2018, 21, 108−121. (b) Wang, Z.; Ge, L.;
REFERENCES Li, M.; Lin, R.; Wang, H.; Zhu, Z. Orientated growth of copper-based
(1) Paris, J. L.; Mannaris, C.; Cabañas, M. V.; Carlisle, R.; Manzano, MOF for acetylene storage. Chem. Eng. J. 2019, 357, 320−327.
M.; Vallet-Regí, M.; Coussios, C. C. Ultrasound-mediated cavitation- (c) Akhbari, K.; Morsali, A. Modulating methane storage in anionic
enhanced extravasation of mesoporous silica nanoparticles for nano-porous MOF materials via post-synthetic cation exchange
controlled-release drug delivery. Chem. Eng. J. 2018, 340, 2−8. process. Dalton Trans. 2013, 42, 4786−4789. (d) Akhbari, K.;
(2) Pundir, S.; Badola, A.; Sharma, D. Sustained release matrix Morsali, A. Needle-like hematite nano-structure prepared by directed
technology and recent advance in matrix drug delivery system: a thermolysis of MIL-53 nano-structure with enhanced methane storage
review. Int. J. Drug Res. Technol. 2013, 3, 12−20. capacity. Mater. Lett. 2015, 141, 315−318.
(3) (a) Hobson, J. J.; Edwards, S.; Slater, R. A.; Martin, P.; Owen, (11) (a) Mirzadeh, E.; Akhbari, K. Synthesis of nanomaterials with
A.; Rannard, S. P. Branched copolymer-stabilised nanoemulsions as desirable morphologies from metal−organic frameworks for various
new candidate oral drug delivery systems. RSC Adv. 2018, 8, 12984− applications. CrystEngComm 2016, 18, 7410−7424. (b) Moeinian, M.;
12991. (b) Rai, V. K.; Mishra, N.; Yadav, K. S.; Yadav, N. P. Akhbari, K. Solid-state synthesis of zinc oxide nano-structures with
Nanoemulsion as pharmaceutical carrier for dermal and transdermal similar morphologies to their precursors and metal−organic frame-
drug delivery: Formulation development, stability issues, basic works topology. J. Iran. Chem. Soc. 2016, 13, 547−552. (c) Moeinian,
considerations and applications. J. Controlled Release 2018, 270, M.; Akhbari, K. How the guest molecules in nanoporous Zn (II)
203−225. metal-organic framework can prevent agglomeration of ZnO nano-
(4) (a) Dimatteo, R.; Darling, N. J.; Segura, T. In situ forming particles. J. Solid State Chem. 2015, 225, 459−463.
injectable hydrogels for drug delivery and wound repair. Adv. Drug (12) (a) Li, Y.; Xie, M.; Zhang, X.; Liu, Q.; Lin, D.; Xu, C.; Xie, F.;
Delivery Rev. 2018, 127, 167−184. (b) Hamedi, H.; Moradi, S.; Sun, X. Co-MOF nanosheet array: A high-performance electro-
Hudson, S. M.; Tonelli, A. E. Chitosan based hydrogels and their chemical sensor for non-enzymatic glucose detection. Sens. Actuators,
applications for drug delivery in wound dressings: A review. B 2019, 278, 126−132. (b) Masih, D.; Chernikova, V.; Shekhah, O.;
Carbohydr. Polym. 2018, 199, 445−460. Eddaoudi, M.; Mohammed, O. F. Zeolite-like Metal−Organic

H https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX
Inorganic Chemistry pubs.acs.org/IC Article

Framework (MOF) Encaged Pt (II)-Porphyrin for Anion-Selective models of drug release. Strategies to Modify the Drug Release from
Sensing. ACS Appl. Mater. Interfaces 2018, 10, 11399−11405. Pharmaceutical Systems 2015, 63−86.
(13) (a) Karimi Alavijeh, R.; Beheshti, S.; Akhbari, K.; Morsali, A. (23) Brozek, C. K.; Michaelis, V. K.; Ong, T.-C.; Bellarosa, L.;
Investigation of reasons for metal−organic framework’s antibacterial Lopez, N. r.; Griffin, R. G.; Dincǎ, M. Dynamic DMF binding in
activities. Polyhedron 2018, 156, 257−278. (b) Usefi, S.; Akhbari, K.; MOF-5 enables the formation of metastable cobalt-substituted MOF-
White, J. Sonochemical synthesis, structural characterizations and 5 analogues. ACS Cent. Sci. 2015, 1, 252−260.
antibacterial activities of biocompatible Copper (II) coordination (24) Ahmed, M.; Abdul Qadir, M.; Imtiaz Shafiq, M.; Muddassar,
polymer nanostructures. J. Solid State Chem. 2019, 276, 61−67. (c) Jo, M.; Hameed, A.; Nadeem Arshad, M.; Asiri, A. M. Curcumin:
J. H.; Kim, H.-C.; Huh, S.; Kim, Y.; Lee, D. N. Antibacterial activities Synthesis optimization and in silico interaction with cyclin dependent
of Cu-MOFs containing glutarates and bipyridyl ligands. Dalton kinase. Acta Pharm. 2017, 67, 385−395.
Trans. 2019, 48, 8084−8093. (25) Bhattacharjee, S. DLS and zeta potential−what they are and
(14) (a) Han, Y.; Liu, W.; Huang, J.; Qiu, S.; Zhong, H.; Liu, D.; what they are not? J. Controlled Release 2016, 235, 337−351.
Liu, J. Materials LettersMaterials & DesignJournal of Porous Materials (26) Lei, B.; Wang, M.; Jiang, Z.; Qi, W.; Su, R.; He, Z. Constructing
Journal of Solid State Chemistry. Pharmaceutics 2018, 10, 271. Redox-Responsive Metal−Organic Framework Nanocarriers for
(b) Chen, X.; Tong, R.; Shi, Z.; Yang, B.; Liu, H.; Ding, S.; Wang, X.; Anticancer Drug Delivery. ACS Appl. Mater. Interfaces 2018, 10,
Lei, Q.; Wu, J.; Fang, W. MOF Nanoparticles with Encapsulated 16698−16706.
Autophagy Inhibitor in Controlled Drug Delivery System for (27) George, P.; Das, R. K.; Chowdhury, P. Facile microwave
Antitumor. ACS Appl. Mater. Interfaces 2018, 10, 2328−2337. synthesis of Ca-BDC metal organic framework for adsorption and
controlled release of Curcumin. Microporous Mesoporous Mater. 2019,
(c) Javanbakht, S.; Pooresmaeil, M.; Namazi, H. Green one-pot
281, 161−171.
synthesis of carboxymethylcellulose/Zn-based metal-organic frame-
(28) (a) Hall, J. N.; Bollini, P. Structure, characterization, and
work/graphene oxide bio-nanocomposite as a nanocarrier for drug
catalytic properties of open-metal sites in metal organic frameworks.
delivery system. Carbohydr. Polym. 2019, 208, 294−301. (d) Abanades
React. Chem. Eng. 2019, 4, 207−222. (b) Alivand, M. S.; Shafiei-
Lazaro, I.; Forgan, R. S. Application of zirconium MOFs in drug Alavijeh, M.; Tehrani, N. H. M. H.; Ghasemy, E.; Rashidi, A.;
delivery and biomedicine. Coord. Chem. Rev. 2019, 380, 230−259. Fakhraie, S. Facile and high-yield synthesis of improved MIL-101
(15) Almásǐ , M.; Zeleňaḱ , V.; Palotai, P.; Beňová, E.; Zeleňaḱ ová, A. (Cr) metal-organic framework with exceptional CO2 and H2S uptake;
Metal-organic framework MIL-101 (Fe)-NH2 functionalized with the impact of excess ligand-cluster. Microporous Mesoporous Mater.
different long-chain polyamines as drug delivery system. Inorg. Chem. 2019, 279, 153−164. (c) Shin, J.; Kim, M.; Cirera, J.; Chen, S.;
Commun. 2018, 93, 115−120. Halder, G. J.; Yersak, T. A.; Paesani, F.; Cohen, S. M.; Meng, Y. S.
(16) (a) Zheng, M.; Liu, S.; Guan, X.; Xie, Z. One-step synthesis of MIL-101 (Fe) as a lithium-ion battery electrode material: a relaxation
nanoscale zeolitic imidazolate frameworks with high curcumin loading and intercalation mechanism during lithium insertion. J. Mater. Chem.
for treatment of cervical cancer. ACS Appl. Mater. Interfaces 2015, 7, A 2015, 3, 4738−4744.
22181−22187. (b) Priyadarsini, K. The chemistry of curcumin: from (29) Zhang, Y.; Wang, L.; Liu, L.; Lin, L.; Liu, F.; Xie, Z.; Tian, H.;
extraction to therapeutic agent. Molecules 2014, 19, 20091−20112. Chen, X. Engineering Metal−Organic Frameworks for Photoacoustic
(17) Bruneau, M.; Bennici, S.; Brendle, J.; Dutournie, P.; Limousy, Imaging-Guided Chemo-/Photothermal Combinational Tumor Ther-
L.; Pluchon, S. Systems for stimuli-controlled release: Materials and apy. ACS Appl. Mater. Interfaces 2018, 10, 41035−41045.
applications. J. Controlled Release 2019, 294, 355−371. (30) Kalita, H.; Prashanth Kumar, B.N.; Konar, S.; Tantubay, S.; Kr.
(18) Yang, M.; Tang, J.; Ma, Q.; Zheng, N.; Tan, L. High activity Fe- Mahto, M.; Mandal, M.; Pathak, A. Sonochemically synthesized
MIL-101 solid acid catalyst for acetalization of aldehydes with biocompatible zirconium phosphate nanoparticles for pH sensitive
methanol and enamination of β-dicarbonyl compounds. J. Porous drug delivery application. Mater. Sci. Eng., C 2016, 60, 84−91.
Mater. 2015, 22, 1345−1350. (31) Akulov, K.; Simkovitch, R.; Erez, Y.; Gepshtein, R.; Schwartz,
(19) (a) Tiwari, A.; Singh, A.; Garg, N.; Randhawa, J. K. Curcumin T.; Huppert, D. Acid effect on photobase properties of curcumin. J.
encapsulated zeolitic imidazolate frameworks as stimuli responsive Phys. Chem. A 2014, 118, 2470−2479.
drug delivery system and their interaction with biomimetic environ- (32) (a) Basu, B. Release kinetics studies of pimozide from buccal
ment. Sci. Rep. 2017, 7, 12598. (b) Li, Y.; Zheng, Y.; Lai, X.; Chu, Y.; mucoadhesive patches. J. Pharm. Res. 2011, 4, 1806−1808.
Chen, Y. Biocompatible surface modification of nano-scale zeolitic (b) Nabipour, H.; Soltani, B.; Ahmadi Nasab, N. Gentamicin Loaded
imidazolate frameworks for enhanced drug delivery. RSC Adv. 2018, Zn2 (bdc)2 (dabco) Frameworks as Efficient Materials for Drug
8, 23623−23628. Delivery and Antibacterial Activity. J. Inorg. Organomet. Polym. Mater.
(20) (a) Ke, F.; Yuan, Y.-P.; Qiu, L.-G.; Shen, Y.-H.; Xie, A.-J.; Zhu, 2018, 28, 1206−1213. (c) Dash, S.; Murthy, P. N.; Nath, L.;
J.-F.; Tian, X.-Y.; Zhang, L.-D. Facile fabrication of magnetic metal− Chowdhury, P. Kinetic modeling on drug release from controlled drug
organic framework nanocomposites for potential targeted drug delivery systems. Acta Polym. Pharm. 2010, 67, 217−223.
delivery. J. Mater. Chem. 2011, 21, 3843−3848. (b) Lestari, W. W.; (33) (a) Sun, C. Y.; Qin, C.; Wang, X. L.; Su, Z. M. Metal-organic
Arvinawati, M.; Martien, R.; Kusumaningsih, T. Green and facile frameworks as potential drug delivery systems. Expert Opin. Drug
synthesis of MOF and nano MOF containing zinc (II) and benzen 1, Delivery 2013, 10, 89−101. (b) Horcajada, P.; Serre, C.; Vallet-Regi,
3, 5-tri carboxylate and its study in ibuprofen slow-release. Mater. M.; Sebban, M.; Taulelle, F.; Ferey, G. Metal-organic frameworks as
Chem. Phys. 2018, 204, 141−146. efficient materials for drug delivery. Angew. Chem., Int. Ed. 2006, 45,
(21) Peppas, N. A.; Narasimhan, B. Mathematical models in drug 5974−8.
delivery: How modeling has shaped the way we design new drug (34) Wang, L.; Guan, H.; Wang, Z.; Xing, Y.; Zhang, J.; Cai, K.
delivery systems. J. Controlled Release 2014, 190, 75−81. Hybrid mesoporous−microporous nanocarriers for overcoming
(22) (a) Mohd Amin, M. C. I.; Butt, A. M.; Katas, H. Synergistic multidrug resistance by sequential drug delivery. Mol. Pharmaceutics
effect of pH-responsive folate-functionalized poloxamer 407-TPGS- 2018, 15, 2503−2512.
mixed micelles on targeted delivery of anticancer drugs. Int. J. (35) Nejadshafiee, V.; Naeimi, H.; Goliaei, B.; Bigdeli, B.; Sadighi,
Nanomed. 2015, 10, 1321−1334. (b) Nabipour, H.; Hossaini Sadr, A.; Dehghani, S.; Lotfabadi, A.; Hosseini, M.; Nezamtaheri, M. S.;
Amanlou, M.; et al. Magnetic bio-metal−organic framework nano-
M.; Rezanejade Bardajee, G. Release behavior, kinetic and
composites decorated with folic acid conjugated chitosan as a
antimicrobial study of nalidixic acid from [Zn2 (bdc)2 (dabco)]
promising biocompatible targeted theranostic system for cancer
metal-organic frameworks. J. Coord. Chem. 2017, 70, 2771−2784.
treatment. Mater. Sci. Eng., C 2019, 99, 805−815.
(d) Gouda, R.; Baishya, H.; Qing, Z. Application of mathematical
models in drug release kinetics of carbidopa and levodopa ER tablets.
J. Dev. Drugs 2017, 6, 1000171. (c) Bruschi, M. L. Mathematical

I https://dx.doi.org/10.1021/acs.inorgchem.9b02756
Inorg. Chem. XXXX, XXX, XXX−XXX

You might also like