Adhikari 2016

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

DOI: 10.1002/cphc.

201501012 Articles

Smart Approach for In Situ One-Step Encapsulation and


Controlled Delivery of a Chemotherapeutic Drug using
Metal–Organic Framework–Drug Composites in Aqueous
Media
Chandan Adhikari and Anjan Chakraborty*[a]

Controlled release of an anticancer drug, doxorubicin (dox), nal stimuli including change to acidic pH and the presence of
from metal–organic framework (MOF)–drug composites is biocompatible liposomes for a period of more than 72 h.
demonstrated under different external stimuli. 1,3,5-Benzenetri- Steady-state fluorescence spectroscopy is used to monitor the
carboxylic acid (H3BTC) is used as an organic ligand, and iron drug release as a function of time and confocal laser scanning
acetate and zinc nitrate are used as metal sources to synthe- microscopy is used to unravel the post-release fate of doxoru-
size Fe–BTC and Zn–BTC MOFs, which are known to be bio- bicin in the presence of liposomes. It is found that drug release
compatible. The in situ formation of MOF–drug composites rate is higher for the Zn–BTC–dox composite than for the Fe–
demonstrates high drug loading capacity compared to conven- BTC–dox composite. This is attributed to the stronger binding
tional methods. The present methodology is devoid of any between dox and Fe-BTC than that between dox and Zn–BTC.
extra steps for loading the drug after synthesis. Moreover, the This study highlights a novel approach for the preparation of
drug loading is also independent of pore size of the MOF as MOF–drug composites in an aqueous medium for future bio-
the drug molecules are embedded inside the MOF during their medical applications.
in situ formation. The drug release was monitored under exter-

1. Introduction

Metal–organic frameworks (MOFs), because of their robustness, high temperatures, toxic solvents, longer reaction times (in
tunable structure, high functionality, high surface area, wide most cases days to weeks), difficulty in scaled-up synthesis
range of pore size, and high porosity, have attracted research- and, of course, need to use sophisticated instruments. The
ers from different branches of science during recent years and toxic solvents used in conventional methods need to be re-
MOFs are used for important applications in various fields.[1, 2] moved carefully before any biomedical applications. To the
MOFs have been used in the different fields of science and in best of our knowledge, there are only a few reports to date
research areas including gas storage, catalysis, gas sensing, that describe the synthesis of MOFs at room temperature.[36–39]
contrast agents, and gas separation.[3–11] In addition to these However, those room-temperature MOF preparations have the
applications, recently MOFs have been used in biomedical ap- drawbacks that they use several highly toxic solvents including
plications for their amphiphilic nature, easily adjustable func- N,N-dimethylformamide, N,N-diethylformamide, and chloro-
tional groups in the framework, possible fine tuning of the form, and require nearly 2–7 days to react.[36–39] These methods
pore size, and so forth, and several reports describe the load- are neither environmentally friendly nor economically viable,
ing and delivery of different drugs.[12–22] Although MOFs have thus limiting their industrial applications. Therefore, if a synthe-
been known for more than four decades, it is only in the last sis is achieved just by using only metal ions and organic li-
15 years that scientists have paid great attention to them.[23] gands in reasonably good yield, and at room temperature in
A large number of MOFs have been synthesized in recent one step without using toxic solvents, then many of the prob-
years according to conventional methodologies, for example, lems associated with the synthesis of MOFs would be solved.
solvothermal, hydrothermal, mechanochemical and electro- Although iron- and zinc-based biodegradable MOFs were
chemical methods.[24–35] These processes have the advantage of synthesized previously using polycarboxylate ligands, they
yielding highly crystalline structures, but suffer from the use of emerged from the use of the conventional methodologies de-
scribed earlier.[14, 40–45] Even the hydrothermal methodology for
[a] C. Adhikari, Dr. A. Chakraborty the synthesis of iron- and zinc-based biodegradable MOFs
Discipline of Chemistry using water as a solvent suffers from the need for high tem-
Indian Institute of Technology Indore
peratures, long reaction times and difficulties in scaled-up syn-
Indore, Madhya Pradesh (India)
E-mail: anjanc@iiti.ac.in thesis, which limits their industrial application.[25] By keeping in
Supporting Information for this article is available on the WWW under mind the high impact of these iron- and zinc-based biocom-
http://dx.doi.org/10.1002/cphc.201501012. patible MOFs in medicinal chemistry, as well as industrial as-

ChemPhysChem 2016, 17, 1070 – 1077 1070 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles
pects, we hereby report a novel approach for the synthesis of
two important biocompatible MOFs at room temperature
using water as a solvent. The use of water removes the possi-
bility of many kinds of toxicity related to solvents and renders
this method greener than previous methods. The room tem-
perature, scaled-up synthesis marks it as more economical and
affords opportunities for industrial applications. Thus, the nov-
elty of our methodology lies in the simple preparation and
short reaction times at room temperature, with the prospect
of easy scale-up. In Table 1, we summarize the advantages of
our method compared to conventional methods.

Table 1. Comparison of the conventional methods of MOF synthesis with


those developed in this work.

Conventional methods[24–35] Method used in this manuscript


1) Need high temperatures 1) Room temperature reaction
(100–200 8C)
2) Uses toxic organic solvents 2) Only water was used
3) Work-up is needed (sometimes 3) No work-up, only centrifugation
this work-up is complex in nature) is needed to collect the product
4) Need more time 4) Only 3–8 h Scheme 1. Structures of doxorubicin and the lipids with which the lipo-
(in general 2–7 days) somes used in this study were made.
5) Need pressurized sealed 5) No need for special conditions
conditions or equipment
6) Yields are 60–80 % 6) 70–80 % yield
7) Highly crystalline product 7) No crystallinity, amorphous in
nature
8) Difficulty in scale up 8) Easily scalable

In addition to their synthesis, we also describe the use of


these MOFs as carriers of a widely used anticancer drug, doxo-
rubicin (dox, Scheme 1). Although MOFs have been used for
the encapsulation of different drugs, it is still a major challenge
to encapsulate significant amounts of larger-sized drug mole-
cules inside MOFs as post-synthetic drug loading is highly de-
pendent on the pore size of the MOF. Here, we propose a new
synthetic route to prepare MOF–drug composites in situ
during the synthesis of the MOF in an aqueous medium
(Scheme 2). This method removes the need for any extra steps
for drug encapsulation after synthesis, and drug loading is in-
dependent of the pore size of the MOF. The methodology also
enables high drug loading (in the case of Zn–BTC, the drug
loading is … 83 % and in the case of Fe–BTC, drug loading is
… 92 %; H3BTC = 1,3,5-benzenetricarboxylic acid).
Scheme 2. Preparation of Fe–BTC and Zn–BTC MOFs at room temperature
Here, we demonstrated the controlled release of dox from
using water as a solvent. The structures of the MOFs depicted here are not
MOFs by using different external stimuli. The encapsulation of the exact single-crystal structure.
doxorubicin and other anticancer drugs such as topotecan, bu-
sulfan, and others, inside MOFs has been studied recently and
the stability of MOF–dox has been studied at physiological In this study, we investigated the effect of pH change on
pH.[46–51] There are several reports that describe successful en- drug release. We also investigated the efficiency of MOF–dox
capsulation and release of different small molecules (e.g. caf- composites as drug delivery systems upon contact with lipo-
feine) from MOFs under the control of different triggering somes, which mimic the cell membranes. The fate of dox in
agents.[15, 52–57] Our group recently studied encapsulation and the presence of liposomes was determined by using confocal
controlled release of dox from zeolitic imidazole frameworks laser scanning microscopy (CLSM) imaging. We were able to
under different external stimuli.[58] encapsulate and control the release of the drug using a com-

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1071 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles
pletely green route, that is, in situ synthesis of MOF–dox com- the dox/zinc nitrate solution with continuous stirring. Stirring was
posites in an aqueous medium. We hope the results from this continued for 8 h at room temperature. The product was collected
study help the future development of on-demand drug deliv- by centrifugation, washed several times with water and dried in an
oven at 100 8C before being used in further experiments (yield:
ery systems and afford opportunities for the industrial applica-
70 %).
tion of MOFs.

Calculation of Encapsulation Efficiency


Experimental Section
The encapsulation efficiency (EE) of dox in both MOFs was calculat-
Materials ed by using a standard equation [Eq. (1)].[47] The EE for Fe–BTC was
Doxorubicin, iron acetate dihydrate, zinc nitrate hexahydrate, 1,3,5- found to be 92 %, whereas the EE for Zn–BTC was 83 %. The EEs
benzenetricarboxylic acid, mesoporous silica, 1,2-dimyristoyl-sn- were comparable to other values reported in the literature.[47] The
glycero-3-phosphocholine (DMPC), 1,2-dimyristoyl-sn-glycero-3- higher EE of Fe–BTC compared to Zn–BTC can be explained based
phosphoglycerol (DMPG), sodium dihydrogen phosphate, and diso- on the binding efficiency of dox to the MOFs.
dium hydrogen phosphate were purchased from Sigma–Aldrich
encapsulated drug ½mg¤
(see Scheme 1 for the structure of lipids and dox). All the reagents EE ½%¤ ¼   100 ð1Þ
drug solution ½mg¤
were used without further purification.

Preparation of Zn–BTC MOF Preparation of Liposomes


Zinc nitrate hexahydrate (5 g, 16.81 mmol) was placed in a round- Liposomes were prepared according to a previously reported pro-
bottom flask and dissolved in water (50 mL). In another flask, 1,3,5- tocol.[59] In brief, an aqueous buffer solution (pH 7.4) was added to
benzenetricarboxylic acid (H3BTC, 1.6 g, 7.614 mmol) was dispersed a round-bottom flask and the temperature was kept above the
in water (50 mL) with sonication for 15 min. Then, the H3BTC sus- phase-transition temperature of the lipids. Then, the required
pension was added to the zinc nitrate solution with continuous amounts of lipids were dissolved in ethanol (less than 1 % v/v) and
stirring. Stirring was continued for 8 h at room temperature to the lipid solution was rapidly injected into the buffer solution at
obtain the optimum yield. Then, the product was collected by cen- above the phase-transition temperature.
trifugation, washed with water and dried in an oven at 100 8C
before being used in further experiments (yield: 72 %).
In Vitro Drug Release Study under Acidic pH

Preparation of Fe–BTC MOF In vitro drug release was studied using fluorescence spectroscopy.
A known amount of solid MOF–dox composite was immersed into
Iron(II) acetate dihydrate (5 g, 28.90 mmol) was added to a round- a phosphate buffer solution (pH … 4.0) and the supernatant was
bottom flask and dissolved in water (50 mL). In another flask, collected over a certain interval of time. The fluorescence emission
H3BTC (1.6 g, 7.614 mmol) was dispersed in water (50 mL) with of the supernatant was measured periodically. The gradual increase
sonication for 15 min. Then, the H3BTC suspension was added to in fluorescence intensity gave a clear indication that the drug mol-
the iron acetate solution with continuous stirring. Stirring was con- ecules were released from the MOF–drug composite.
tinued for 3 h at room temperature to obtain the optimum yield.
Then, the product was collected by centrifugation, washed with
water and dried in an oven at 100 8C before being used in further In Vitro Drug Release Study in the Presence of Liposomes
experiments (yield: 83 %).
Solid MOF–drug composite was immersed into a liposome prepa-
ration composed of DMPG/DMPC (1:9) and the fluorescence inten-
In Situ Doxorubicin Loading in Fe–BTC MOF sity of the supernatant was measured periodically to monitor the
drug release. The uptake of drug molecules by liposomes was also
Dox was added to water (50 mL) to give a concentration of shown by CLSM.
0.1 mg mL¢1. Next, iron(II) acetate dihydrate (5 g, 28.90 mmol) was
added and fully dissolved by stirring for 15 min. In another flask,
H3BTC (1.6 g., 7.614 mmol) was dispersed in water (50 mL) with Instrumentation
sonication for 15 min. Then, the H3BTC suspension was added to
We characterized MOFs and MOF–dox composites by powder
the dox/iron acetate solution with continuous stirring. Stirring was
X-ray diffraction, scanning electron microscopy and infrared spec-
continued for 3 h. The product was collected by centrifugation,
troscopy. Powder X-ray diffraction was performed in an automated
washed several times with water and dried in an oven at 100 8C
multipurpose Rigaku SmartLab X-ray diffractometer. The X-ray gen-
before being used in further experiments (yield: 82 %).
erator was a 3 kW sealed tube X-ray generator (maximum voltage
60 kV, maximum current 50 mA, with a Cu target). Field-emission
scanning electron microscopy (FE-SEM) studies were conducted on
In Situ Doxorubicin Loading in Zn–BTC MOF
a Zeiss Supra 55 electron microscope. Steady-state absorption
Dox was added to water (50 mL) to give a concentration of spectra were recorded on a Varian Cary 100 UV/Vis spectrometer.
0.1 mg mL¢1. Next, zinc nitrate hexahydrate (5 g, 16.81 mmol) was IR spectra were measured on a Tensor 27 FTIR spectrometer
added and fully dissolved by stirring for 15 min. In another flask, (Bruker). Thermogravimetric analysis (TGA) was performed on
H3BTC (1.6 g., 7.614 mmol) was dispersed in water (50 mL) with a TGA/DSC1 instrument from Mettler Toledo (Switzerland) under ni-
sonication for 15 min. Then, the H3BTC suspension was added to trogen flow at a heating rate of 5 8C per minute. The drug released

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1072 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles
was monitored by steady-state fluorescence spectroscopy using To monitor the in situ loading of the drug and its release
a Horiba Jobin Yvon Fluoromax-4p FM-100 spectrofluorimeter. The from the MOFs, we used UV/Vis and fluorescence spectroscopy.
samples were excited at 480 nm. For the time-resolved studies, we To confirm that dox was embedded inside the MOF, we mea-
used a picosecond time-correlated single-photon counting (TCSPC)
sured absorption and emission spectra for both the initial dox
system from IBH (model Fluorocube-01-NL). The experimental
solution in water (diluted fivefold as the initial concentration
setup for TCSPC has been described elsewhere.[60, 61] The samples
were excited at 480 nm using a picosecond diode laser and the was high) and supernatant after the reaction. Figure 2 a shows
decays were collected at 600 nm. The repetition rate was 5 MHz. the UV/Vis spectra for dox in an aqueous medium and after
The signals were collected at magic-angle (54.708) polarization forming composites with MOFs. It is clear from Figure 2 a that
using a photomultiplier tube (TBX-07C) as the detector. The full after formation of the composite, the absorption of dox was
width at half-maximum of the decay function was around 140 ps. diminished almost to zero for Fe–BTC MOF. This indicates that
Uptake of drugs by liposomes were studied by CLSM on an Olym- most of the dox molecules are embedded in Fe–BTC. Con-
pus IX83 microscope (multiple Ar laser, XY LSM). The wavelength
versely, in the case of Zn–BTC–dox, a few dox molecules re-
of the excitation laser was 488 nm. The morphologies of the MOFs
mained in solution and thus showed a weak absorption at
were also studied by CLSM and bright-field imaging.
490 nm. Figure 2 b shows the fluorescence spectra for dox in
aqueous medium (diluted fivefold) and after forming compo-
sites with MOFs. The data also indicates that after the drug is
embedded in Fe–BTC, there is no free dox remaining in the su-
2. Results and Discussion
pernatant and hence there is no emission at 590 nm for the su-
After the synthesis, we characterized the MOFs and MOF–drug pernatant. However, in the case of Zn–BTC–dox, even after
composites by SEM. The images show that the morphology re- composite formation takes place, few dox molecules remain in
mains the same for both MOF and MOF–drug composite solution, which showed weak emission at 590 nm. The result
(Figure 1). implies that Fe–BTC has a stronger binding affinity [EE around
Both MOFs were stained with the well-known dye rhodami- 92 % using Eq. (1)] for dox as compared to Zn–BTC [EE around
ne B for the CLSM and bright-field studies. The excitation 83 %].
wavelength was set at 559 nm and the emission was collected The FTIR spectroscopy of the powders of Fe–BTC, Zn–BTC
at 625 nm. Figure 1 e and f show the CLSM and bright-field and Fe–BTC–dox and Zn–BTC–dox composites are shown in
images, respectively, for the Zn–BTC MOF, and Figure 1 g and h Figure 1 c. The bands centered at 1550 and 1422 cm¢1 corre-
are the CLSM and the bright-field images, respectively, for the spond to the C=O and C¢O stretching frequencies, respective-
Fe–BTC MOF. The images are in good agreement with the SEM ly, for the free carboxylate groups of H3BTC. The bands cen-
images. From CLSM, bright-field and SEM studies, it is clear tered at 1610 and 1380 cm¢1 correspond to C=O and C¢O
that the size of the MOFs are close to one micrometer. group of carboxylic acid after coordination to the metal
center.[62, 63] The bands centered at 1324, 1390, 1615, 1720,
2525, and 3000–3500 cm¢1 (broad band) correspond to ether
C¢O and carboxylic acid C¢O, C=C, C=O and O¢H, and O¢H of
dox, respectively.
TGA was performed to gain information about the thermal
stability of the MOFs and drug-embedded MOFs. Figure 3 a
shows that Fe–BTC and Zn-BTC MOFs are stable up to 300 and
285 8C, respectively. The higher thermal stability of Fe–BTC
over Zn–BTC can be explained by the electronic configuration
of the metal ions. Metal complexes with d10 electronic configu-
ration, such as Zn2 + , are less stable than complexes with d6
configuration such as Fe2 + due to the ligand-field stabilization
energy. Drug-embedded MOFs show the same thermal stability
as blank MOFs. At the beginning, for both the MOF–dox com-
posites, the small weight loss (around 10 %) at 150 8C takes
place due to the thermal decomposition of dox. A sharp
weight loss (75 %) was observed for both the MOF after 350 8C.
This weight loss indicates thermal decomposition of the MOF
into its corresponding metal oxide in that temperature range.
Figure 3 b shows the powder X-ray diffraction (PXRD) pattern
of synthesized MOFs and MOF–drug composites. The sharp
peak of the PXRD pattern indicates a uniform size distribution
of the particles. The PXRD pattern of pure Fe–BTC and Zn–BTC
match with those reported previously.[62, 63] Figure 3 b also
Figure 1. SEM images of a) Zn–BTC, b) Zn–BTC–dox, c) Fe–BTC, and d) Fe–
BTC–dox (scale bars = 1 mm). e) CLSM and f) bright-field image of Zn–BTC. shows that there was no change in the PXRD pattern of MOFs
g) CLSM and h) bright-field image of Fe–BTC. after forming composites with drug molecules. We did not

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1073 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles

Figure 2. a) Absorption spectra for dox in water (diluted fivefold), dox-embedded Fe–BTC MOF and dox-embedded Zn–BTC MOF. b) Fluorescence emission
spectra of dox, dox-embedded Fe–BTC MOF and dox-embedded Zn-BTC MOF. d) IR spectra of Fe–BTC, dox-embedded Fe–BTC MOF, Zn–BTC, and dox-embed-
ded Zn–BTC MOF; d) expanded IR spectra of Fe–BTC–dox and Zn–BTC–dox.

Figure 3. a) TGA curves for blank MOFs and dox-embedded MOFs. b) PXRD patterns of blank MOFs and dox-embedded MOFs. The emission spectra of dox in
phosphate buffer (pH … 4.0) at varying times from 0–72 h for c) Fe–BTC–dox and d) Zn–BTC–dox. Inset: plots of intensity versus time are shown. Exponential
fitting of % drug release versus time under acidic conditions for e) Zn–BTC–dox and f) Fe–BTC–dox.

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1074 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles
choose a rigorous structural characterization of the MOFs as f ¼ að1 ¢ expð¢kt ÞÞ ð3Þ
the structures of both MOFs have been studied extensively by
other groups and single-crystal structures are available in the
literature.[40, 63] where k is the rate constant of release and a is the pre-expo-
So, the MOFs prepared in the current methodology were nential factor, which is a measure of the extent of the release
well characterized by PXRD, TGA, IR and SEM, CLSM and of contents (Figure 3 e and f).
bright-field spectroscopy. From the UV/Vis, fluorescence, IR, Thus, the fitting yielded rate constants of around 0.03 and
TGA and PXRD data it is clear that MOFs were formed and that 0.01 h¢1 for Zn–BTC and Fe–BTC MOF, respectively. The lower
drug molecules were embedded inside MOF successfully. rate constant for Fe–BTC might be due to the stronger binding
After the characterization, we shifted our focus to the re- ability of dox with the Fe–BTC MOF than with the Zn–BTC
lease of drugs under the stimulus of changes in pH. We chose MOF, as indicated by UV/Vis and fluorescence results. There-
pH … 4 and 5 (data not shown) for our study as cancer cells are fore, the drug molecules are released from the Zn–BTC frame-
acidic (pH 4–6) in nature.[64, 65] To investigate drug release, the work at a faster rate than Fe–BTC framework.
solid MOF–drug composites (500 mg) were immersed in phos- After pH stimuli, we investigated the effect of biomimetic
phate-buffer solution (pH … 4.0, 10 mL) and the release of drug systems such as liposomes on MOF–dox composites as lipo-
molecules was monitored by fluorescence emission of the su- somes are known to mimic cell membranes and are considered
pernatants. The drug release was monitored for a period of to be a true biomimetic system. We used negatively charged
72 h. Figure 3 c and d shows that there is a monotonic incre- liposomes composed of two lipids (DMPC/DMPG, 8:2). As the
ment in fluorescence intensity at 590 nm as time increases. pKa of dox is 8.2, it remains mostly positively charged under
This gradual increase in intensity with time implies the release the experimental conditions (pH … 7.4) used.[46] Therefore, it is
of drug molecules from the framework. At acidic pH the car- likely that dox will bind strongly with the negatively charged
boxylate anions are protonated and the coordination between liposomes. To establish this fact, we studied the interaction of
carboxylic acid group and metal center breaks down, which re- dox with bare liposomes (see Figure S1 and Table S1 in the
sults in disruption of the MOF leading to drug release. Fig- Supporting Information). Figure 4 a and b shows the release of
ure 3 c shows that release of drug was complete by a period of drugs upon contact with liposomes. It is clear from Figure 4 a
72 h in the case of Fe–BTC–dox, whereas Zn–BTC–dox took and b that there is an almost 1.6-fold increase in fluorescence
60 h to completely release its drug. The percentage of drug re- intensity in the case of Fe–BTC–dox and the increment was
lease was estimated using Equation (2): 2.8-fold in the case of Zn–BTC–dox. This is also attributed to
the stronger binding affinity of Fe–BTC with dox compared to
F t ¢F t¼0 Zn–BTC. Notably, the intensity at zero time in the presence of
%release ¼ ð2Þ liposomes is much higher compared to that in buffer solution.
F ¢F t¼0
t¼1

This fact indicates that in the presence of liposomes, dox mole-


cules absorbed on the surface of the MOFs immediately re-
where F0 is the fluorescence intensity at zero time, Ft is the lease from the composites due to electrostatic interactions
fluorescence intensity at time t and Ft = 1 is the maximum in- with the liposomes.
tensity if all dox molecules are released from the composite. The post-release fate of dox upon addition of liposomes to
The rate constant was obtained by fitting to a single-expo- MOF–dox composites was also studied by CLSM. CLSM images
nential rate equation following first-order kinetics [Eq. (3)]: (Figure 5) reveal that there was no emission from blank lipo-

Figure 4. The emission spectra of doxorubicin in the presence of liposomes at times varied from 0–72 h. a) Fe–BTC–dox, b) Zn–BTC–dox.

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1075 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles
[3] J. L. Rowsell, O. M. Yaghi, Angew. Chem. Int. Ed. 2005, 44, 4670 – 4679;
Angew. Chem. 2005, 117, 4748 – 4758.
[4] M. Dincă, J. R. Long, Angew. Chem. Int. Ed. 2008, 47, 6766 – 6779; Angew.
Chem. 2008, 120, 6870 – 6884.
[5] J. Zhuang, C.-H. Kuo, L.-Y. Chou, D.-Y. Liu, E. Weerapana, C.-K. Tsung, ACS
Nano 2014, 8, 2812 – 2819.
[6] B. Chen, S. Ma, E. J. Hurtado, E. B. Lobkovsky, C. Liang, H. Zhu, S. Dai,
Inorg. Chem. 2007, 46, 8705 – 8709.
[7] B. Chen, S. Xiang, G. Qian, Acc. Chem. Res. 2010, 43, 1115 – 1124.
[8] H.-L. Jiang, Y. Tatsu, Z.-H. Lu, Q. Xu, J. Am. Chem. Soc. 2010, 132, 5586 –
5587.
[9] H.-L. Jiang, Q. Xu, Chem. Commun. 2011, 47, 3351 – 3370.
[10] J. Lee, O. K. Farha, J. Roberts, K. A. Scheidt, S. T. Nguyen, J. T. Hupp,
Chem. Soc. Rev. 2009, 38, 1450 – 1459.
[11] C.-H. Kuo, Y. Tang, L.-Y. Chou, B. T. Sneed, C. N. Brodsky, Z. Zhao, C.-K.
Figure 5. CLSM images of a) blank liposomes, b) MOF–dox-liposomes, Tsung, J. Am. Chem. Soc. 2012, 134, 14345 – 14348.
c) zoomed image of liposomes after uptake of dox. [12] J. Della Rocca, D. Liu, W. Lin, Acc. Chem. Res. 2011, 44, 957 – 968.
[13] P. Horcajada, T. Chalati, C. Serre, B. Gillet, C. Sebrie, T. Baati, J. F. Eubank,
D. Heurtaux, P. Clayette, C. Kreuz, Nat. Mater. 2010, 9, 172 – 178.
[14] P. Horcajada, C. Serre, M. Vallet-Reg†, M. Sebban, F. Taulelle, G. F¦rey,
somes. As liposome is added to MOF–dox composites, the Angew. Chem. Int. Ed. 2006, 45, 5974 – 5978; Angew. Chem. 2006, 118,
drug release takes place and an intense signal was obtained 6120 – 6124.
from solution. Furthermore, it is revealed that the drug mole- [15] J. An, S. J. Geib, N. L. Rosi, J. Am. Chem. Soc. 2009, 131, 8376 – 8377.
[16] N. Li¦dana, A. Galve, C. Rubio, C. T¦llez, J. Coronas, ACS Appl. Mater. In-
cules appear to be in the core of the liposome and were uni-
terfaces 2012, 4, 5016 – 5021.
formly distributed in the bulk as a result of release from MOF– [17] I. Imaz, J. Hernando, D. Ruiz-Molina, D. Maspoch, Angew. Chem. Int. Ed.
dox composites. This confirmed the successful uptake of dox 2009, 48, 2325 – 2329; Angew. Chem. 2009, 121, 2361 – 2365.
molecules into liposomes. [18] I. Imaz, M. Rubio-Mart†nez, L. Garc†a-Fern‚ndez, F. Garc†a, D. Ruiz-Molina,
J. Hernando, V. Puntes, D. Maspoch, Chem. Commun. 2010, 46, 4737 –
4739.
[19] S. Devautour-Vinot, C. Martineau, S. Diaby, M. Ben-Yahia, S. Miller, C.
3. Conclusions Serre, J. Phys. Chem. C 2013, 117, 11694 – 11704.
[20] D. Cunha, C. Gaudin, I. Colinet, P. Horcajada, G. Maurin, C. Serre, J.
In conclusion, we report a simple, environmentally benign
Mater. Chem. B 2013, 1, 1101 – 1108.
method for the synthesis of two highly biocompatible MOFs [21] R. Babarao, J. Jiang, J. Phys. Chem. C 2009, 113, 18287 – 18291.
and drug-embedded MOFs in a one-step, room temperature [22] S. Devautour-Vinot, G. Maurin, C. Serre, P. Horcajada, D. Paula da Cunha,
reaction. The drug-embedded MOFs were found to release the V. Guillerm, E. de Souza Costa, F. Taulelle, C. Martineau, Chem. Mater.
2012, 24, 2168 – 2177.
drug in the presence of a biomimetic system such as lipo-
[23] H. Li, M. Eddaoudi, M. O’Keeffe, O. M. Yaghi, Nature 1999, 402, 276 – 279.
somes. This is the first time that liposomes have been used to [24] S. J. James, Chem. Soc. Rev. 2003, 32, 276 – 288.
release dox from a dox-embedded MOF. We also demonstrated [25] A. U. Czaja, N. Trukhan, U. Mìller, Chem. Soc. Rev. 2009, 38, 1284 – 1293.
the successful release of the drug from MOF–drug composites [26] J. R. Long, O. M. Yaghi, Chem. Soc. Rev. 2009, 38, 1213 – 1214.
using low-acidity conditions over a period of three days for the [27] Z. Wang, S. M. Cohen, Chem. Soc. Rev. 2009, 38, 1315 – 1329.
[28] M. Eddaoudi, J. Kim, N. Rosi, D. Vodak, J. Wachter, M. O’Keeffe, O. M.
first time. The novelty of the present work is in its facile and in- Yaghi, Science 2002, 295, 469 – 472.
dustrially amenable synthesis of the MOFs and drug-embed- [29] K. Schlichte, T. Kratzke, S. Kaskel, Microporous Mesoporous Mater. 2004,
ded MOFs and in the slow delivery of dox using external stim- 73, 81 – 88.
uli. We hope this study will afford new opportunities in the [30] A. Pichon, A. Lazuen-Garay, S. L. James, CrystEngComm 2006, 8, 211 –
214.
field of biomaterials science in the near future.
[31] J. A. Rood, B. C. Noll, K. W. Henderson, Inorg. Chem. 2006, 45, 5521 –
5528.
[32] O. M. Yaghi, H. Li, J. Am. Chem. Soc. 1995, 117, 10401 – 10402.
Acknowledgements [33] J. L. C. Rowsell, O. M. Yaghi, Microporous Mesoporous Mater. 2004, 73,
3 – 14.
[34] O. M. Yaghi, M. O’Keeffe, N. W. Ockwig, H. K. Chae, M. Eddaoudi, J. Kim,
The authors would like to thank Sophisticated Instrument Centre
Nature 2003, 423, 705 – 714.
(SIC), IIT Indore for providing the facilities and infrastructure. We [35] A. Czaja, E. Leung, N. Trukhan, U. Mìller in Metal-Organic Frameworks:
are grateful to the powder X-ray diffraction (P-XRD) facility at the Applications from Catalysis to Gas Storage (Ed.: D. Farrusseng), Wiley-
SIC, IIT Indore. C.A. acknowledges financial support from IIT VCH, Weinheim, 2011, pp. 337 – 352.
Indore. [36] J. R. Hunt, O. M. Yaghi, Tetrahedron 2008, 64, 8553 – 8557.
[37] M. Gerardo, I. Oliver, Y. Maxim, J. Gunnar, J. P¦rez-Ram†rez, CrystEng-
Comm 2013, 15, 9885 – 9892.
Keywords: doxorubicin · drug delivery · liposomes · metal– [38] J. L. Zhuang, D. Ceglarek, S. Pethuraj, A. Terfort, Adv. Funct. Mater. 2011,
21, 1442 – 1447.
organic frameworks · pH
[39] L. Huang, H. Wang, J. Chen, Z. Wang, J. Sun, D. Zhao, Y. Yan, Micropo-
rous Mesoporous Mater. 2003, 58, 105 – 114.
[1] N. W. Ockwig, O. Delgado-Friedrichs, M. O’Keeffe, O. M. Yaghi, Acc. [40] P. Horcajada, S. Surble, C. Serre, D.-Y. Hong, Y.-K. Seo, J.-S. Chang, J.-M.
Chem. Res. 2005, 38, 176 – 182. Greneche, I. Margiolaki, G. Ferey, Chem. Commun. 2007, 2820 – 2822.
[2] M. Eddaoudi, D. B. Moler, H. Li, B. Chen, T. M. Reineke, M. O’Keeffe, O. M. [41] V. Agostoni, R. Anand, S. Monti, S. Hall, G. Maurin, P. Horcajada, C. Serre,
Yaghi, Acc. Chem. Res. 2001, 34, 319 – 330. K. Bouchemal, R. Gref, J. Mater. Chem. B 2013, 1, 4231 – 4242.

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1076 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Articles
[42] A. G. M‚rquez, A. Demessence, A. E. Platero-Prats, D. Heurtaux, P. Horca- [53] Q. Hu, J. Yu, M. Liu, A. Liu, Z. Dou, Y. Yang, J. Med. Chem. 2014, 57,
jada, C. Serre, J. S. Chang, G. Ferey, V. A. de la PeÇa-O’Shea, C. BoissiÀre, 5679 – 5685.
Eur. J. Inorg. Chem. 2012, 32, 5165 – 5174. [54] L.-N. Duan, Q. – Q. Dang, C.-Y. Han, X.-M. Zhang, Dalton Trans. 2015, 44,
[43] Y.-K. Seo, J. W. Yoon, J. S. Lee, U.-H. Lee, Y. K. Hwang, C.-H. Jun, P. Horca- 1800 – 1804.
jada, C. Serre, J.-S. Chang, Microporous Mesoporous Mater. 2012, 157, [55] N. Li¦dana, A. Galve, C. Rubio, C. T¦llez, J. Coronas, ACS Appl. Mater. In-
137 – 145. terfaces 2012, 9, 5016 – 5021.
[44] L. Xu, E.-Y. Choi, Y.-U. Kwon, Inorg. Chem. Commun. 2008, 11, 1190 – [56] L. Nuria, L. Pablo, G. Alejandro, T. Carlos, C. Joaqu†n, J. Mater. Chem. B
1193. 2014, 2, 1144 – 1151.
[45] L. Luo, K. Chen, Q. Liu, Y. Lu, T. Okamura, G. C. Lv, Y. Zhao, W. Y. Sun, [57] P. Lorena, P. Gr¦gory, A. Steven, C. Joaqu†n, Org. Biomol. Chem. 2015, 13,
Cryst. Growth Des. 2013, 13, 2312 – 2321. 1724 – 1731.
[46] R. Anand, F. Borghi, F. Manoli, I. Manet, V. Agostoni, P. Reschiglian, R. [58] C. Adhikari, A. Das, A. Chakraborty, Mol. Pharm. 2015, 12, 3158 – 3166.
Gref, S. Monti, J. Phys. Chem. B 2014, 118, 8532 – 8539. [59] R. Thakur, A. Das, A. Chakraborty, J. Photochem. Photobiol. B 2014, 130,
[47] M. R. di Nunzio, V. Agostoni, B. Cohen, R. Gref, A. Douhal, J. Med. Chem. 122 – 131.
2014, 57, 411 – 420. [60] R. Thakur, A. Das, A. Chakraborty, Phys. Chem. Chem. Phys. 2012, 14,
[48] T. Chalati, P. Horcajada, P. Couvreur, C. Serre, M. Ben Yahia, G. Maurin, R. 15369 – 15378.
Gref, Nanomedicine 2011, 6, 1683 – 1695. [61] C. Adhikari, A. Das, A. Chakraborty, ChemPhysChem 2015, 16, 866 – 871.
[49] A. C. McKinlay, R. E. Morris, P. Horcajada, G. Ferey, R. Gref, C. Couvreur, C. [62] M. Tong, D. Liu, Q. Yang, S. Devautour-Vinot, G. Maurinb, C. Zhonga, J.
Serre, Angew. Chem. Int. Ed. 2010, 49, 6260 – 6266; Angew. Chem. 2010, Mater. Chem. A 2013, 1, 8534 – 8537.
122, 6400 – 6406. [63] X. Huang, Y. Chen, Z. Lin, X. Ren, Y. Song, Z. Xu, X. Dong, X. Li, C. Hu, B.
[50] R. Canioni, C. Roch-Marchal, F. S¦cheresse, P. Horcajada, C. Serre, M. Wang, Chem. Commun. 2014, 50, 2624 – 2627.
Hardi-Dan, G. F¦rey, J. M. GrenÀche, F. Lefebvre, J. Chang, Y. Hwang, O. [64] N. W. Read, K. Sugden, Crit. Rev. Ther. Drug Carrier Syst. 1998, 4, 221 –
Lebedev, S. Turnerf, G. Van Tendeloo, J. Mater. Chem. 2011, 21, 1226 – 263.
1233. [65] T. T. Kararli, Biopharm. Drug Dispos. 1995, 16, 351 – 380.
[51] V. Agostoni, T. Chalati, P. Horcajada, H. Willaime, R. Anand, N. Semira-
moth, T. Baati, S. Hall, G. Maurin, H. Chacun, K. Bouchemal, C. Martineau,
F. Taulelle, P. Couvreur, C. Rogez-Kreuz, P. Clayette, S. Monti, C. Serre, R.
Gref, Adv. Healthcare Mater. 2013, 2, 1630 – 1637. Manuscript received: November 9, 2015
[52] D. Cunha, M. B. Yahia, S. Hall, S. R. Miller, H. Chevreau, E. Elkam, G. Accepted Article published: January 11, 2016
Maurin, P. Horcajada, C. Serre, Chem. Mater. 2013, 25, 2767 – 2776. Final Article published: February 16, 2016

ChemPhysChem 2016, 17, 1070 – 1077 www.chemphyschem.org 1077 Ó 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like