You are on page 1of 20

www.advancedsciencenews.com www.advmat.

de

Metal–Organic Framework (MOF)-Based Drug/Cargo

Review
Delivery and Cancer Therapy
Ming-Xue Wu and Ying-Wei Yang*

biocompatible, i.e., they should be non-


Metal–organic frameworks (MOFs)—an emerging class of hybrid porous toxic or have low toxicity and should be
materials built from metal ions or clusters bridged by organic linkers—have easily degraded by the body’s metabo-
attracted increasing attention in recent years. The superior properties of lism. Furthermore, the release of drugs
in drug-loaded vehicles is expected to be
MOFs, such as well-defined pore aperture, tailorable composition and struc-
effectively controlled. As a result, exten-
ture, tunable size, versatile functionality, high agent loading, and improved sive efforts have been devoted to applying
biocompatibility, make them promising candidates as drug delivery hosts. various controlled drug release systems
Furthermore, scientists have made remarkable achievements in the field of in cancer chemotherapy.[7] More impor-
nanomedical applications of MOFs, owing to their facile synthesis on the tantly, scientists are currently focusing
on the development of multimodal
nanoscale and alternative functionalization via inclusion and surface chem-
treatment systems to satisfy an ever-
istry. A brief introduction to the applications of MOFs in controlled drug/ growing demand for efficient therapeutic
cargo delivery and cancer therapy that have been reported in recent years is strategies.[8]
provided here. Hitherto, metal–organic frameworks
(MOFs) have been designed via diverse
synthetic strategies[9] and studied for
1. Introduction many applications, including gas storage and separations,[11]
[10]

catalysis,[12] sensing,[13] nonlinear optics,[14] an increasing


Cancer is a threat to the health of human beings that causes number of potential biomedical applications,[9e,10a,15] and many
millions of deaths annually. Chemotherapy is still the domi- others.[7d,9a,15b,16] Figure 1 summarizes the main synthetic
nant treatment method. However, traditional direct adminis- methodologies of MOFs and their applications. In recent years,
tration of therapeutic drugs to patients is no longer applicable MOFs (where “MOF” encompasses all possible dimension
due to intrinsic limitations, including undesirable side effects, scales) have received extensive attention in the development
poor pharmacokinetics and poor biodistribution.[1] Thus, var- of drug delivery and cancer theranostic platforms, particularly
ious efforts have been made toward the development of drug MOFs in the nanoscale regime.[9a,d,15b,17] Compared with tradi-
vehicles for controllable drug release that achieve reduced tional drug carriers, mentioned above, MOFs possess the fol-
side effects and enhanced therapeutic efficacy.[2] Accordingly, lowing obvious advantages:[7b,18] Firstly, their versatile struc-
during the last few decades, many different types of nanocar- tures provide MOFs with diverse morphologies, compositions,
riers have been developed for this purpose, such as inorganic sizes, and chemical properties, which endow them with multi-
mesoporous silica, quantum dots, metal nanoparticles (NPs), functionalities and stimuli-responsive drug controlled release.
and organic micelles, liposomes, and dendrimers.[3] How- Moreover, it is notable that the modification of such materials
ever, all nanocarriers have their own limitations in bioappli- has no significant alteration of their desirable physicochemical
cations; liposomes, micelles, and dendrimers usually suffer properties, i.e., MOF-based materials still maintain control-
from low loading capacities,[4] and inorganic porous materials lable size, shape, and high uniformity.[19] Secondly, large sur-
have undesirable toxicity and unacceptable degradability.[5] An face areas and high porosities facilitate MOFs with high loading
ideal drug-loading system should possess the following basic capacity. Thirdly, weak coordination bonds ensure MOFs are
requirements: carriers should have a large loading capacity; biodegradable. In short, these desirable properties enable
carriers should be nanoscale in order to facilitate the release MOFs to be promising platforms for drug delivery, clinical
of drugs by intravenous administration;[6] carriers must be tumor therapy, and other disease therapies.
The scope of this review will highlight developments of
MOFs in the fields of controllable drug release and efficacious
M.-X. Wu, Prof. Y.-W. Yang
International Joint Research Laboratory of treatment of cancer, including drug nanocarriers and cancer
Nano-Micro Architecture Chemistry (NMAC) theranostic platforms composed of individual MOFs (Table 1),
College of Chemistry stimuli-responsive MOFs (Table 2), and multifunctional MOFs
Jilin University (Table 3), respectively. The essential demands of MOFs for bio-
2699 Qianjin Street, Changchun 130012, China
E-mail: ywyang@jlu.edu.cn
applications are also addressed. Finally, the prospects and chal-
lenges for MOFs have been extensively discussed. In contrast
DOI: 10.1002/adma.201606134 to other excellent reviews that discuss the bioapplications of

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (1 of 20) 1606134
www.advmat.de www.advancedsciencenews.com

MOFs from a broad perspective, this review differs in that it


Review

will focus on highlighting the applications of MOFs, from indi- Ming-Xue Wu received her
vidual nanocarriers to multifunctional cancer theranostic plat- B.Sc. (2014) in chemistry
forms, and provides an outlook for the development of cancer from Shandong University
theranostic platforms for MOFs and their potential clinical of Technology, China. She
applications. became an M.Sc. student
at Jilin University in 2014,
where she performed
2. Individual MOFs as Drug Nanocarriers research under the direc-
tion of Professor Qiong Jia.
MOFs intrinsically possess large surface areas, highly ordered In 2016, she started her
porosities, and well-defined structures, which endow these Ph.D. in the research group of
materials with the capability of loading and releasing dif- Professor Ying-Wei Yang. Her
ferent cargos, especially therapeutic agents. In 2006, Férey research focuses on the fabrication of stimuli-responsive
and co-workers first proposed that MOFs have potential in MOF-based drug nanocarriers to realize efficient cancer
drug delivery, owing to their considerable loading capaci- treatment.
ties and controlled release behavior.[20] They built up two Cr-
based MOFs, denoted MIL-100 and MIL-101, from trimers of
Ying-Wei Yang received his
metal octahedra and di- or tricarboxylic acids, which exhib-
B.Sc. (2000) and Ph.D. (2005)
ited remarkable IBU adsorption with 0.35 g g–1 of IBU/dehy-
from Nankai University. From
drated MIL-100 and 1.4 g g–1 of IBU/dehydrated MIL-101.
2005 to 2010, he gained
The release kinetics of IBU were studied in simulated body
postdoctoral training at ASU
fluid at 37 °C, indicating that a complete release of IBU from
(with John C Chaput), UCLA
MIL-100 occurred after 3 days, and from MIL-101 after 6 days.
(with Sir Fraser Stoddart) and
Compared with a similar material, MCM-41, these two hybrid
UCI (with Zhibin Guan). In
solids showed comparable IBU dosages and kinetics in sim-
2011, he became an Associate
ulated body fluid; especially MIL-101(Cr), which may be due
Professor in the College of
to its larger cage sizes and stronger interactions with IBU.
Chemistry at Jilin University
Notably, this work developed a new method of MOF-based
and was promoted to Full
drug delivery.
Professor in 2014. His research interests include multi-
Taking into account the toxicity of chromium compounds,
functional organic–inorganic hybrid nanomaterials, organic
some low toxicity MOFs have been applied for the delivery of
supramolecular chemistry, and stimuli-responsive (bio)
drug molecules. For instance, varieties of available Fe-based
polymers.
MOFs offer many possibilities to achieve an adequate controlled
release of various pharmacological molecules.[9a,69] In 2008, the
much less toxic iron analogue, MIL-53(Fe), composed of tere-
phthalate anions and trans chains of Fe (III) octahedra, was
used to adsorb IBU by impregnation of the
solids in a hexane IBU-containing solution
with loading capacity of 20 wt%.[21] Then, the
delivery of IBU was performed in simulated
body fluid at 37 °C under continuous stir-
ring. A complete delivery of IBU took around
21 days, which was proved as zero-order
kinetics. Importantly, this work highlighted
for the first time the unique properties of
MOFs in terms of adapting their pore open-
ings to the dimensions of drugs, to optimize
drug-matrix interactions. Besides, porous
MIL-100 (Fe) was used as a nanocarrier of
antitumor drug doxorubicin (DOX), whereby
a dosage of 9 wt% was received with complete
release after 2 weeks.[7a] Moreover, MIL-100
(Fe) or iron aminoterephthalate have been
applied for encapsulating highly hydrophilic
nucleoside analogues, antivirals cidofovir
(CDV), and azidothimidine triphosphate
(AZT-TP), with high loading capacities of
Figure 1. Schematics showing the synthesis and applications of MOFs. 42 wt%, 29 wt%, and 21 wt%, respectively.[7a]

1606134 (2 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

Table 1. Examples of individual MOFs as drug nanocarriers.

Review
Individual MOFs as drug nanocarriers
MOFs Major components Drug/cargo Loading percentage Targeted cell lines Ref.
[wt%]
MIL-100/101 (Cr) Trimers of chromium octahedral, 1,3,5-BTC/1,4-BDC IBU 25.8/58 − [20]
MIL-53 (Fe) Fe3+, 1,4-BDC IBU 20 – [21]
Zn-TATAT Zn2+, TATAT 5-Fu 33.3 − [22]
Zn-H2BDP_X Zn2+, H2BDP_X (X = H, NO2, NH2, OH) Mitroxantrone, PAPTA_C − J774 [23]
Zn-CDDB Zn2+, H2CDDB 5-Fu 53.3 HepG2, MDA-MB-435s [24]
Zn2(1,4-bdc)2(dabco) Zn2+, 1,4-BDC, 1,4-diazabicyclo [2.2.2] octane IBU 15 HuH7 [25]
UiO NMOF ZrCl4, amino-TPDC Cisplatin prodrug, siRNA 12.3 ± 1.2 Ovarian cancer cells [26]
Nano-UiO-66-2-NH2 ZrCl4, 1,4-BDC_NH2 5-Fu 27 − [27]
UiO-66_X Zirconium polyhedral, 1,4-BDC_X (X = H, Br, NH2) Caffeine 22.4 ± 3.4 (X = H) − [28b]
21.2 ± 0.7 (X = Br)
13.2 ± 0.2 (X = NH2)
bMOF-1/100 Zinc adenine-SBU, BPDC/Azo-BPDC Etilefrine hydrochloride 10 ± 0.7 (bMOF-1) − [29]
bMOF-4/102 10.2 ± 0.6 (bMOF-4)
9.6 ± 0.8 (bMOF-100)
10.9 ± 0.9(bMOF-102)
MIL-100 (Fe) Fe3+, 1,3,5-BTC DOX 9 − [7a]
Nucleoside 42 −
CDV 29 −
AZT-TP 21 Human peripheral blood
mononuclear cells

Zn-based MOFs have also been reported as nanocar- completion. Results showed that the loaded drug was released
riers due to the low toxicity of zinc ion. For example, Wang over a long time ranging from 49 to 80 days, although an initial
et al. have reported a chiral Zn-based MOF constructed from burst release was observed. Cao et al. also synthesized a water-
5,5′ 5′′-(1,3,5-triazine-2,4,6-triyl) tris (azanediyl) triisophtha- stable Zn-based MOF–Zn8(O)2(CDDB)6(DMF)4(H2O)] (CDDB =
late (TATAT) and Zn2+ for the delivery of anticancer drug 4,4′-(9-H carbazole-3,6-diyl) dibenzoic acid) and 5-Fu was adsorbed
5-fluorouracil (5-Fu).[22] Adsorption of 5-Fu was achieved by into the host MOF.[24] The drug delivery capacity of the synthe-
soaking the prepared MOF in 5-Fu containing methanol solu- sized MOF was around 53.3 wt% with a delivery time of 3 days.
tions, and the hydrogen bonding interactions between 5-Fu Usually, there are two methods for drug-loading: one method
and the MOF ensured a loading capacity of 0.5 g g−1. Drug is a two-step encapsulation of drugs by immersing the pre-
was released from the drug-loaded MOF by dialyzing the as- pared MOFs nanocarriers into drug-containing solutions; the
prepared drug vehicle against phosphate buffered saline (PBS) other is a one-pot encapsulation of drugs. In addition to the
solution (pH 7.4) at room temperature, and the drug release two-step encapsulation of drugs mentioned above, Morsali and
process lasted for a week. Barea et al. have reported a series co-workers reported the first example of in situ synthesis of a
of isoreticular MOFs composed of Zn2+ and some function- drug-loaded MOF at room temperature.[25] IBU-loaded MOF
alized organic linkers 1,4-bis(1H-pyrazol-4-yl)-2-X-benzene (Zn2(1,4-bdc)2(dabco))n was studied as a drug delivery system
(H2BDP_X; X = H, NO2, NH2, OH).[23] Following this, two with loading capacity of 15 wt% and a progressive release over
antitumor drugs, mitroxantrone and [Ru(p-cymene)Cl2(pta)] 12 days.
(RAPTA-C, pta = 1,3,5-triaza-7-phospaadamantane), were Recently, a series of zirconium-based MOFs, especially
encapsulated in the porous Zn-based MOFs. An et al. reported UiO-66 (UiO for Oslo University),[70] have received increasing
that four types of adenine-based MOFs (bMOFs) were selected attentions.[71] These are composed of Zr6O4(OH)4 secondary
for delivery of etilefrine hydrochloride.[29] Among them, building units (SBUs) and dicarboxylate bridging ligands.
bMOF-1 and bMOF-100 were constructed from a zincadeninate UiO-66 MOFs are desirable candidates for drug delivery because
secondary building unit (SBU) and biphenyl-4,4′-dicarboxylic of their two octahedral (≈11 Å) and tetrahedral (≈8 Å) cages,
acid (BPDC), and bMOF-4 and bMOF-102 were built from the nontoxicities, and excellent stabilities.[28] Serre et al. studied the
same SBU with azobenzene-4,4′-dicarboxylic acid (Azo-BPDC). rationality of a variety of biocompatible MOFs as nanocarriers
Drug-loading was accomplished by soaking the bMOFs combined with experimental and computational systematic
in solutions of etilefrine hydrochloride in ethanol, after exploration.[28b] Cubic UiO-66 solids were constructed from zir-
which the drug was released into simulated body fluid until conium oxoclusters and terephthalate anions bearing different

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (3 of 20) 1606134
www.advmat.de www.advancedsciencenews.com
Review

Table 2. Examples of stimuli-responsive MOFs for drug delivery

Single-stimulus-responsive MOFs for drug delivery


Stimuli MOFs Major components Drug/cargo Loading Targeted cell lines Ref.
percentage [wt%]
pH Fe(bbi) Fe2+, bbi DOX 40 Hela cells [30]
PAA@ZIF-8 PAAS, Zn2+, HMeIM DOX 65.5 MCF-7 [31]
UiO-66 ZrCl4, 1,4-BDC AL 51.5 MCF-7, HepG2 [32]
MG-Gd-pDBI Gd3+, pDBI DOX 12 U937 [33]
ZIF-8 Zn2+, 2-methylimidazolate CPT 2 MCF-7 [34]
ZIF-8 Zn2+, 2-methylimidazolate DOX 20 MCF-7, MDA-MB-231, [35]
MDA-MB-468
ZJU-101 Zirconium, BPYDC Diclofenac sodium 35.3 PC12 [36]
PCN-221 ZrCl4, TCPP MTX 28.6 PC12 [37]
Magnetic field Fe3O4/Cu3(BTC)2 2+
Cu , 1,3,5-BTC, Fe3O4 nanorods NIM 14.5-16.7 − [38]
γ-Fe2O3@MIL-53(Al) Al3+, 1,4-BDC, γ-Fe2O3 IBU 10 − [39]
Ions bioMOF-1 Zinc adeninate-SBUs, Procainamide HCl 18 − [40]
biphenyldicarboxylate
MOF-74-Fe(III) Fe2+, Ibuprofen anions 15.9 PC12 [41]
2,5-dioxido-1,4-benzenedicarboxylate
Temperature UiO-66-PNIPAM ZrCl4, 1,4-BDC_NH2, PNIPAM Resorufin, Caffeine, − − [42]
Procainamide
ZJU-64/ ZJU-64-CH3 Zn2+, adenine, terphenyldicarboxylic MTX 13.5/10.6 PC12 [43]
acid
Pressure ZJU-800 ZrOCl2·8H2O, F-H2PDA Diclofenac sodium 58.8 PC12 [44]
Multiple-stimuli-responsive MOFs for drug delivery
pH, competitive agent CP5-capped Zn2+, 1,4-BDC_NH2, 4,4′,4′′-ben- DOX 3.4 HEK293 [45]
UMCM-1-NH-Py zene-1,3,5-triyl-tribenzoic acid, CP5, Py
Zn2+, thermo CP5-capped ZrCl4, 1,4-BDC_NH2, CP5, quaternary 5-Fu 1.5 HEK293 [46]
UiO-66-NH2 ammonium salt
Ca2+, pH, thermo CP5-capped ZrCl4, 1,4-BDC_NH2, CP5, quaternary 5-Fu 3.1 HEK293 [47]
UiO-66-NH2 ammonium salt
pH, redox PEG-RGD-β-CD- PEG-1900, k(ad)RGDS, β-CD-S-S-BCN, DOX 13.4 Hela cells, COS7 cells [48]
SS-MIL-101 MIL-101-N3(Fe)
Amantadine, light β-CD-capped ZrCl4,2′-p-tolyldiazenyl-1,1′:4,4′- RhB − − [49]
UiO-68-azo terphenyl-4,4′′-dicarboxylic acid, β-CD
pH, liposomes Fe-BTC, Zn-BTC Fe2+/Zn2+, 1,3,5-BTC DOX − − [50]

functional groups on the aromatic benzyl ring (UiO-66_X for Ling et al. introduced a continuous flow microreactor tech-
X = H, Br, or NH2). Caffeine encapsulation was performed by nology for preparation of downsized UiO-66 NPs.[27] High
simple immersion, indicating that UiO-66 were promising car- surface-to-volume ratio of the continuous flow microreactor
riers of caffeine. In another study, Lin et al. reported the first facilitated efficient heat transfer, leading to fast nucleation of
example of UiO MOFs for co-delivery of cisplatin and pooled UiO-66 NPs in the nanoscale, meanwhile the growth of UiO-
siRNAs to enhance their therapeutic efficacy by overcoming 66 MOFs could be controlled by residence time. Furthermore,
drug resistance in ovarian cancer cells.[26] UiO MOF was pre- the obtained NH2-functionalized nano-UiO-66-2 (nano-UiO-
pared with ZrCl4 and aminotriphenyldicarboxylic acid (amino- 66-2-NH2) was used for loading 5-Fu, with a loading capacity of
TPDC) bridging ligands, in which a cisplatin prodrug and 27 wt%. This work provided a facile strategy for the preparation
siRNA were sequentially loaded by encapsulating and coordi- of MOFs for bioapplications.
nating to metal sites on the MOF surfaces (Figure 2a). Green
fluorescence was clearly observed in cells incubated with
siRNA/UiO-cis, but not in those treated with siRNA/UiO or 3. Stimuli-Responsive MOFs for Drug Delivery
UiO-cis, indicating that the developed siRNA/UiO-cis induce
cell apoptosis (Figure 2b–d). All these results demonstrated Smart materials have aroused extensive attentions especially in
that such a co-delivery method would enhance the chemother- the field of bioapplications.[72] Among them, stimuli-responsive
apeutic efficacy of cisplatin in vitro (Figure 2e). Interestingly, MOFs have been appealing candidates for controllable drug

1606134 (4 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

Table 3. Examples of multifunctional MOFs for cancer theranostics.

Review
Multifunctional MOFs for cancer theranostics
Polymer-coated MOFs
Composites Major components Drug/cargo Functionality Targeted cell lines Ref.
MIL-101-NH2(Fe)@Silica@c(RGDfk) Fe3+, 1,4-BDC_NH2, silica shell, Br-BODIPY, Cisplatin prodrug Chemotherapy, Optical HT-29 [51]
ESCP prodrug, c(RGDfk) imaging, Targeting
Mn-bisphosphonate@Lipid@peg-AA Mn2+, zoledronic acid, DOPA, DOPC, Zoledronate Chemotherapy, MRI, MCF-7, AsPC-1 [52]
DSPE-PEG2k, cholesterol, anisamide Targeting
Hep-MIL-100(Fe) Fe3+, 1,3,5-BTC, Rhodamine-labeled heparin Caffeine Stealth property, Low J774.A1, HL60 [53]
inflammatory responses,
Chemotherapy
UiO-66/67@PCL-TPGS Poly(ε-caprolactone)-tocopheryl poly(ethylene Cisplatin, Taxol Chemotherapy, Sustained U-87MG, HSC-3 [54]
glycol)-succinate copolymer, UiO-66, UiO-67 release, Reduced toxicity
Magnetic core–shell MOFs
Fe3O4@UiO-66 ZrCl4, 1,4-BDC, Fe3O4 DOX Magnetic separation, MRI, HeLa cells, 3T3 cells [55]
Chemotherapy
Fe3O4@ZIF-90 Zn2+, imidazolate-2-carboxyaldehyde, rat serum 5-Fu MRI, Magnetic thermal − [56]
albumin, PVP, Fe3O4 effect, Chemotherapy
Fe3O4@IRMOF-3@FA-RITC Zn2+, 1,4-BDC_NH2, Fe3O4, PVP, folic acid, Paclitaxel Chemotherapy, MRI, HeLa cells, NIH3T3 [57]
rhodamine B isothiocyanate Targeting, Optical imaging
Fe3O4@PAA/AuNCs/ZIF-8 Zn2+, HMeIM, HAuCl4, GSH, PAA, Fe3O4 DOX MRI, CT, Optical imaging, HepG2, H22 [58]
pH responsiveness
Fe3O4@C@MIL-100(Fe) Fe3+, 1,3,5-BTC, Ferrocene, Fe3O4 DHA, Fe(III) Magnetic guidance, MRI, A549, HeLa cells [59]
Optical imaging, pH
responsiveness,
Chemotherapy, Yield ROS
Fe3O4/ZIF-8-Au25 Zn2+, HMeIM, A25(Capt)18−, Fe3O4 − MRI, Targeting, Photo­ L929, HeLa cells [60]
dynamic therapy, Photo-
thermal therapy
Other core–shell MOF nanocomposites
Mn3[Co(CN)6]2@SiO2@Ag Mn2+,K3[Co(CN)6], PVP, SiO2, AgNO3 DOX MRI, TPF imaging, A549, HeLa cells, [61]
Photothermal therapy, HepG2
Chemotherapy, Light
responsiveness
PB@MIL-100(Fe) Fe3+, 1,3,5-BTC, PVP, K3[Fe(CN)6] Artemisinin MRI, Optical imaging, Photo­ HeLa cells [62]
thermal therapy, Chemo-
therapy, pH responsiveness
Mn-IR825@PDA-PEG Mn2+, IR825, polydopamine, PEG − MRI, Photothermal therapy 293T, HeLa cells, [63]
A549, 4T1
Hf-TCPP-PEG HfCl4, TCPP, C18PMH-PEG − Photodynamic therapy, 4T1, HeLa cells, [64]
Radiation therapy NIH3T3
Fc-Gd@SiO2(RBITC)-RGD Gd3+, Fc, silica, RBITC, c(RGDfk) − MRI, Targeting U87MG, MCF-7 [65]
UCNP@Fe-MIL-101_NH2@PEG@FAs NaYF4:Yb, Er, PVP, Fe3+,
1,4-BDC_NH2, − UCL property, MRI, Targeting KB cells, MCF-7 [66]
NH2-PEG-COOH, folic acid
Flexible MOF-based theranostic nanoplatforms
Mn∥coordination polymers Mn(ClO4)2, non-steroidal anti- NSAIDs Cell imaging, Drug delivery RAW264.7 [67]
inflammatory drugs (NSAIDs),
1,2-bis(4-pyridyl)ethylene
UiO-PDT ZrCl4, 1,4-BDC, I2-BDP − Photodynamic therapy B16F10, C26, CT26 [68]

release. Typically, stimuli-responsive MOFs can be divided MOFs and multiple-stimuli-responsive MOFs, which are able to
into two categories: single-stimulus- and multiple-stimuli- achieve regulated delivery of loaded drugs upon activation by
responsive MOFs. In this part, we introduce a variety of stim- diverse stimuli, such as pH, magnetic field, ions, temperature,
uli-responsive MOFs, including single-stimulus-responsive light, and pressure.

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (5 of 20) 1606134
www.advmat.de www.advancedsciencenews.com
Review

Figure 2. a) Schematic presentation of siRNA/UiO-cis synthesis and drug-loading. b–d) CLSM images showing cell apoptosis and siRNA (TAMRA-
labeled, red) internalization in SKOV-3 cells after incubation with UiO-cis (b), siRNA/UiO (c), and siRNA/UiO-cis (d) for 24 h. The apoptotic cells
were stained with Alexa Fluor 488 Annexin V conjugate and the nuclei were stained with DAPI. Scale bar represents 10 µm. e) Cytotoxicity of siRNA/
UiO-cis in SKOV-3 cells after 72 h incubation determined by MTS assay. Reproduced with permission.[26] Copyright 2014, American Chemical Society.

3.1. Single-Stimulus-Responsive MOFs for Drug Delivery Consequently, confocal laser scanning microscopy (CLSM)
analysis showed that this pH-sensitive drug release formulation
3.1.1. pH-Responsive MOFs showed a faster drug release in buffer solution of pH value 5.5
than at a neutral pH of 7.4. Such pH-dependent drug nanocar-
Of all porous MOF nanocarriers triggered by external stimuli, riers were taken up by MCF-7 cells through endocytosis and
pH-responsive MOFs are the most widely investigated, espe- were nearly nontoxic to live cells, suggesting their potential
cially for cancer therapy, because of the acidic tumor microen- bioapplications.
vironment and the sensitivity of coordination bonds to external Shi and co-workers also reported a pH-responsive drug car-
pH.[73] Until now, many investigations have involved the pH- rier, UiO-66 MOF, which was applied for alendronate (AL)
responsive properties of MOFs for drug delivery and cancer delivery owing to its natural drug anchorages of Zr–O clus-
treatment. For example, Huang et al. reported an Fe (bbi) MOF, ters.[32] Herein, an ultrahigh dosage of AL, up to 1.06 g AL g−1
constructed from 1,1′-(1,4-butanediyl) bis (imidazole) (bbi) and UiO-66, was obtained due to strong complexation of Zr–O–P
ferrous ions by a deposition method.[30] In situ encapsulation of bonds. Next, the release behavior of AL loaded nanocompos-
DOX was achieved by simply adding DOX into the bbi solution ites was assessed at 37 °C in PBS buffer at pH = 5.5/7.4; the
(Figure 3a) and, as expected, the loaded DOX could be released released amounts within 60 h were 59% and 42.7%, respec-
upon lowering the pH, due to the sensitivity of the coordination tively. Such release behavior may be attributed to protonation
bond to the acidic environment (Figure 3b). Thus, a layer of of the AL at lower pH, which weakens the complexation of
silica was coated on the surface of the MOF to prevent the rapid Zr–O–P bonds. Nevertheless, over a prolonged time the release
decomposition of the hosting material for effective control over rate was higher at pH 7.4 than that at pH 5.5; a unique behavior
the release of DOX. More importantly, folic acid was conjugated that is attributed to the higher stability of UIO-66 under acidic
to the surface of the as-prepared nanocomposites for targeted conditions than under neutral and basic conditions.[70] In vitro
drug delivery (Figure 3c). Consequently, the MOF-based drug cytotoxicity measurements showed that AL-UiO-66 exhibited
vehicle showed pH-dependent progressive release behavior and higher anticancer efficacy than free AL after 48 h of incubation,
good anticancer efficacy. indicating that pH-responsive AL-UiO-66 improved antitumor
Another pH-sensitive nanocarrier, poly(acrylic acid)@zeolitic efficiency and made UiO-66 a promising carrier for AL delivery.
imidazolate framework-8 (PAA@ZIF-8), was prepared by ion- Interestingly, a Gadolinium (III)-based MOF, Gd-pDBI
exchange between Zn2+ and poly(acrylic acid sodium salt) (pDBI = 1,4-bis(5-carboxy-1H- benzimidazole-2-yl)benzene),
(PAAS), followed by reacting with 2-methylimidazole (HMeIM) was proved to be a potential pH-responsive drug vehicle.[33] In
in methanol solution.[31] Notably, the obtained PAA@ZIF-8 this work, Gd-pDBI was transformed to MG-Gd-pDBI upon
nanocarrier exhibited high DOX loading capability (1.9 g DOX mechanical grinding and showed good aqueous dispersibility.
g−1 NPs), which may be attributed to electrostatic interactions It was then used for intravenous injection. The pre-synthe-
between the negatively charged PAA and positively charged sized MG-Gd-pDBI showed higher DOX loading capacity than
DOX, as well as the coordination interaction of Zn(II)-DOX. Gd-pDBI due to its enhanced surface exposure towards DOX.

1606134 (6 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

Review
Figure 3. a) Schematic presentation of DOX/Fe(bbi)@SiO2–FA. b) In vitro pH-responsive release profiles of DOX·HCl from DOX/Fe(bbi) and DOX/
Fe(bbi)@SiO2. c) CLSM imaging showing that folate receptor mediated endocytosis enhanced HeLa cells uptake. Reproduced with permission.[30]
Copyright 2013, Royal Society of Chemistry.

Moreover, about 44% of drug was released from the nano- loaded with 20 wt% of DOX were proved to be efficient nano-
carrier at pH 5 within 5 days, whereas only 22% of loaded carriers for cancer therapy via pH-responsive release due to the
DOX was released at pH 7.4 after 5 days, suggesting that the instability of ZIF-8 under acidic conditions.
obtained MOF possessed pH-responsive drug release. Both Recently, the Qian group described an interesting cationic
in vitro and in vivo experiments demonstrated the practicality nanocarrier, MOF ZJU-101 (ZJU = Zhejiang University), for
of the pre-synthesized MG-Gd-pDBI for DOX release and its delivery of diclofenac sodium, an anionic drug.[36] The cationic
desirable biocompatibility without side effects to other major host material was composed of zirconium and 2,2′-bipyridine-
organ functions. 5,5′-dicarboxylate (BPYDC) with high diclofenac sodium
Tsung et al. reported a pH-responsive ZIF-8 with uniform loading capacity of 0.546 g g–1. Diclofenac sodium was released
70 nm NPs as a theranostic system.[34] Negatively charged faster in the inflamed tissues (pH = 5.4) than in normal tis-
small molecules such as fluorescein camptothecin (CPT) could sues (pH = 7.4), because the ion exchange between anions in
be encapsulated into the ZIF-8 NPs by one-pot encapsulation. the PBS and the drug occurred more frequently at acidic con-
The performance of pH-responsive release was driven by the ditions, which weaken the Coulombic interaction between the
dissociation of the ZIF-8 framework at the acidic designated cationic ZJU-101 and anionic drug. Therefore, pH-sensitive
target. Herein, CPT-ZIF-8 treated cells showed increased cell diclofenac sodium@ZJU-101 was expected to be a promising
death compared with free CPT treated cells. Similarly, Zou anti-inflammatory drug vehicle.
et al. reported a one-pot method for synthesis of ZIFs (ZIF-8 More interestingly, they reported the first example of PCN-
and ZIF-67) to encapsulate organic molecules (anticancer drug 221 MOF as an oral drug carrier.[37] PCN-221 was prepared
DOX and dye molecules, such as rhodamine B, methyl orange, from 5,10,15,20-tetrakis (4-carboxyphenyl) porphyrin (TCPP)
and methylene blue).[35] Research results demonstrated that the and ZrCl4, and then the anticancer drug methotrexate (MTX)
developed one-pot process could be used to encapsulate target was encapsulated by immersing PCN-221 in MTX solution.
organic molecules into the same ZIF crystals. ZIF-8 crystals Subsequently, the MTX-PCN-221 with loading capacity of

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (7 of 20) 1606134
www.advmat.de www.advancedsciencenews.com

0.40 g g–1 was immersed in PBS solution at 37 °C with pH 3.1.3. Ion-Responsive MOFs
Review

values of 7.4 and 2.0, respectively. Experimental results exhib-


ited that ≈40% of MTX was released at pH = 2.0 (around Ion-responsive MOFs have introduced a new pathway for drug
the pH of stomach) after 3 days, whereas 100% of MTX was delivery. In such drug carriers, strong electrostatic interactions
released at pH = 7.4 (close to the intestinal pH). Accordingly, between the drugs and the frameworks control the diffusion
the pH-responsive MTX-PCN-221 was expected to be a prom- and release of drugs. Thus, the strong electrostatic interactions
ising oral drug carrier. between ionic drugs and ionic frameworks have attracted par-
ticular interest because the release of ionic drugs is a chem-
ical stimuli-responsive process taking place only through ion
3.1.2. Magnetically-Responsive MOFs exchange. For instance, the Rosi group fabricated an anionic
Zn8(ad)4(BPDC)6O·2Me2NH2·8DMF·11H2O (bioMOF-1) by
Magnetically responsive systems allow for diversity in drug introducing biphenyldicarboxylic acid to the mixtures of ade-
delivery due to their potential superiorities in magnetic sepa- nine and zinc acetate dihydrate.[40] Such anionic bioMOF-1
ration, magnetic targeting, magnetic resonance imaging was explored to test its capability to storage and release of pro-
(MRI), and magnetic hyperthermia.[74] Among these, mag- cainamide HCl, a cationic antiarrythmia drug. Because of the
netically guided anticancer drug delivery is a unique strategy electrostatic interaction between anions and cations, a loading
to concentrate drug-loaded therapeutic probes in tumor sites dosage of 0.22 g g–1 was achieved and the cations in the bio-
to improve therapeutic efficacy, since its first introduction by logical fluids were expected to trigger the release of procaina-
Watson et al. in the 1960s.[75] Candidate nanocarriers based mide HCl. Therefore, as a controlled experiment, drug-loaded
on MOFs for such a delivery approach are usually core–shell bio-MOF-1 was placed in a PBS buffer (pH = 7.4) and nanopure
NPs, for example Fe3O4 was often used as a magnetic core with water, which verified that drug release could be triggered by
a MOF shell. For instance, Zhang et al. described a targeted buffer cations.
drug vehicle based on a magnetic MOF, i.e., Fe3O4/Cu3(BTC)2 Yang and co-workers developed a cationic drug carrier, MOF-
(HKUST-1, BTC = benzene-1,3,5-tricarboxylate) nanocompos- 74-Fe(III), through the oxidation of neutral MOF-74-Fe(II).[41]
ites.[38] Nimesulide (NIM), an anticancer drug, was loaded in The cationic MOF exhibited around 15.9 wt% of the loading
the magnetic nanocarrier by soaking Fe3O4/Cu3(BTC)2 in the capacity of ibuprofen anions (Figure 4a). Interestingly, the
NIM trichloromethane solution. Magnetic measurements preparation process of MOF could yield hydroxide, resulting in
were carried out, suggesting that Fe3O4/Cu3(BTC)2 possessed the presence of two distinctive Ibu− anions in MOF channels,
desirable magnetic performance for targeted drug delivery and thus, two different release behaviors were observed. Sodium
separation. ibuprofen and the other coordinated free anions were released
In 2014, Guan and co-workers reported a one-step in situ at first, merely because of diffusion or ion exchange between
pyrolysis method for the fabrication of γ-Fe2O3@MOFs. In the drug-encapsulated MOF and PBS solution. The following
their work, γ-Fe2O3@MIL-53(Al) demonstrated its potential release profile of the targeted drug was triggered by phosphate
for controllable magnetic separation and drug release.[39] As anions through competitive adsorption (Figure 4b).
expected, the magnetic nanocomposites exhibited controlled
release behavior in physiological saline at 37 °C; that is,
the encapsulated IBU was completely released after 7 days 3.1.4. Temperature-Responsive MOFs
in three stages: firstly, about 30% of the drug was released
rapidly at the first 3 hours; then, 50% of drug was released Generally, temperature-responsive nanocarriers are materials
during the next 2 days; finally, the residual 20% of drug was that are sensitive to slight temperature changes at the physi-
released over 5 days. These results verified that the magnetic ological temperature of 37 °C. Poly(N-isopropyl acrylamide)
γ-Fe2O3@MIL-53(Al) material was feasible for drug delivery. (PNIPAM) is a promising building block for thermosensitive
Very recently, versatile magnetically responsive MOF-based drug nanocarriers that possess a lower critical solution temper-
biomedical carriers have been developed and are discussed in ature.[76] When the temperature is lowered to below its cloud
detail in 4. point (Tc, around 32 °C), PNIPAM exhibits hydrophilicity and

Figure 4. a) Schematic fabrication process of MOF-74-Fe(II) oxidation to crystal 1 and drug-loading. b) Release profiles of Ibu− in PBS solution under
2.0 and 4.0 MPa, respectively. Reproduced with permission.[41] Copyright 2014, American Chemical Society.

1606134 (8 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

Review
Figure 5. a) Schematic showing the controlled release profiles of UiO-66-PNIPAM. b) Release behavior of drug-loaded UiO-66-PNIPAM in water at
25 °C and 40 °C for seven days. c) Temperature-responsive release behavior of resorufin from UiO-66-PNIPAM in water at 572 nm. Reproduced with
permission.[42] Copyright 2015, Royal Society of Chemistry.

tends to dissolve in water; however, it also forms an aggregate. and ZJU-64-CH3, respectively. The same amount of MTX was
Accordingly, Sada et al. demonstrated a switchable UiO-66- released from MTX-loaded ZJU-64 and ZJU-64-CH3 at 37 °C
PNIPAM nanocarrier (Figure 5a).[42] Resorufin, caffeine, and for 72 h, but only took approximately 1.5 and 6 h at 60 °C, sug-
procainamide were loaded in the nanocarrier by soaking the gesting a potential application of ZJU-64 and ZJU-64-CH3 as
UiO-66-PNIPAM in guest solutions, then the release behaviors temperature-responsive drug carriers.
were assessed at 25 °C or 40 °C (Figure 5b). As expected, the
amount of released drug increased at 25 °C and almost halted
at 40 °C (Figure 5c), which indicated that controlled release was 3.1.5. Pressure-Responsive MOFs
driven by temperature variation.
Recently, the Qian group synthesized two zinc-based MOFs, To avoid the premature release of drugs until they reach at path-
Zn16(ad)8(TP)8O2(H2O)8·4HTP·36DMF·16H2O (ad = adenine, ological tissues, above-mentioned responsive MOFs have been
H2TP = [1,1′:4′,1′′-terphenyl]-4,4′′- dicarboxylic acid for ZJU- developed with prolonged release time and enhanced thera-
64, and 2′,5′-dimethyl-[1,1′:4′,1′′-terphenyl]-4,4′′-dicarboxylic peutic effcacy. In addition to the above commonly mentioned
acid for ZJU-64-CH3).[43] Anticancer drug MTX was encapsu- stimuli, pressure has also been employed for controllable drug
lated into the two MOFs by a simple impregnation method, release. Recently, Qian and co-workers reported a Zr-based MOF
with loading a capacity of 13.45 and 10.63 wt% for ZJU-64 builted from (2E,2E′)-3,3′-(2-fluoro-1,4-phenylene) diacrylic

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (9 of 20) 1606134
www.advmat.de www.advancedsciencenews.com

acid (F-H2PDA) and zirconium cluster with high model drug positively-charged pyridinium (Py) stalks (CP5-capped UMCM-
Review

diclofenac sodium (DS) loading capacity of 58.80 wt% due to 1-NH–Py). In acidic tumor tissues or tumor cells, the gatekeeper
its enhanced polarity and extended organic spacer.[44] The DS- of CP5 tended to be neutral, which weakened the noncovalent
release kinetics of such drug-loaded MOF could be adjusted binding interactions between CP5 and the stalk, leading to
through varying pressure, resulting in a prolonged release time unblocking of the MOF followed by the release of cargo. Upon
between 2–8 days. Such formulation showed a new method for addition of a competitive binding agent (methylviologen salts),
responsive MOF-based drug delivery. which possess a higher binding affinity to CP5 than Py, CP5
was detached from the stalks, which caused the release of cargo.
We integrated pH- and/or competitive binding agent-responsive
3.2. Multiple-Stimuli-Responsive MOFs for Drug Delivery strategies into a single drug nanocarrier (Figure 6), where the
CP5-capped drug nanocarrier showed high encapsulation effi-
The complexity of the human body environment limits ciency, negligible premature release, negligible cytotoxicity,
abilities to precisely deliver drugs in the body using single- desirable biodegradability, and biocompatibility. Further studies
stimulus-responsive MOF drug carriers. However, multiple- of the multiple-stimuli-sensitive nanocarriers based on MOFs
stimuli-responsive MOFs can be used as a better alternative to with CP5 rings as gatekeeper have also been carried out in
improve loading capacities and chemotherapeutic efficiencies. our group. For instance, we have reported another CP5-capped
Pillararenes, one example of rapidly developed supramolecular UiO-66-NH2 5-Fu nanocarrier.[46] In this work, UiO-66-NH2 was
hosts, have been widely investigated since their first discovery modified with positively charged quaternary ammonium salt as
by Ogoshi et al. in 2008. This is due to their desirable proper- stalks (Q) to thread the negatively charged CP5 rings, to form
ties, including their special structure, facile functionalization, [2]pseudorotaxanes as stimuli-responsive supramolecular gates
and desirable host–guest performance.[77] Certainly, this new that regulate drug release. Zn2+ can be a competitive binding
class of macrocycles is promoting the development of supramo- agent to trigger the release of 5-Fu because of a higher binding
lecular chemistry and materials science.[78] Accordingly, many affinity between Zn2+ and CP5. In addition, increasing tempera-
host–guest systems based on pillararenes have been devel- ture could also weaken the noncovalent bonding interactions
oped,[79] in particular, pillararene-based nanovalves for drug between the CP5 and stalks to stimulate the release of drugs.
delivery have recieved increasing attention in recent years.[80] It Therefore, the dual stimuli-responsive drug nanocarrier based
is conceivable that the combination of MOFs with pillararenes on CP5-gated MOF outlines a new opportunity for therapy of
will reveal a new method for the bioapplications of MOFs. central nervous system diseases.
Our research group has devoted much effort to design such Recently, our group developed a triply-responsive theranostic
versatile drug nanocarriers. According to the rapid development hybrid platform based on CP5-gated Zr-MOFs.[47] Positively
of gated materials,[81] multiple-stimuli-responsive drug delivery charged quaternary ammonium salt stalks were tethered on
system (DDS) composed of MOFs and pillararene-based supra- UiO-66-NH2 via post-synthetic modification, and then negatively
molecular nanovalves was developed for the first time by our charged CP5 was encircled on the stalks sitting on the surface
research group, in collaboration with the Wang group.[45] of the MOF through host–guest complexation. As mentioned
UMCM-1-NH2 was used as the host material to store cargoes of above, low pH, increasing temperature, and competitive binding
guest molecules, followed by modification of carboxylatopillar[5] agents could weaken the supramolecular interactions between
arene (CP5) as a dual stimuli-responsive gatekeeper linked by the MOF scaffold and the gating nanovalves of CP5 to control

Figure 6. Schematic illustration of dual-stimuli responsive DDS based on UMCM-1-NH2 NMOF gated by pillararenes. Reproduced under the terms of
the CC-BY-NC-3-0 unported license.[45] Copyright 2014, The Royal Society of Chemistry.

1606134 (10 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

Review
Figure 7. Schematic illustration of triple-stimuli-responsive DDS based on CP5-capped UiO-66. Reproduced with permission.[47] Copyright 2015, Royal
Society of Chemistry.

the targeted release of drugs. Therefore, herein, a triple modal Light-responsive release and competitive binding agent-respon-
therapeutic trigger was integrated in one system (Figure 7). sive release were implemented in the system by tethering β-CD
Controlled release experiments verified that the prepared thera- on the surface of UiO-68-azobenzene (β-CD–capped UiO-68-azo)
peutic platform was promising for potential bone regeneration through supramolecular complexation (Figure 9a). Azobenzene
and bone cancer therapy because lower pH and higher con- groups provided photo-controllable cis/trans isomerizations and
centration of Ca2+ in osteoclasts and tumor cells could stimu- the isomerization of azobenzene from trans to cis promoted the de-
late the release of the therapeutic agents via pH-responsiveness threading of the gating β-CD to regulate cargo release (Figure 9b).
and Ca2+-competitive responsiveness. Furthermore, high tem- Furthermore, a competitive binding agent such as amantadine
peratures could damage and kill cancer cells as well as weaken could also trigger the dissociation of the gatekeeper from the
host–guest interactions to prompt gradually release of the thera- azobenzene stalks due to its higher binding affinity toward β-CD
peutic molecule (5-Fu). Furthermore, the triply-responsive drug (Figure 9c). Rhodamine B was loaded in the dual-stimuli respon-
nanocarrier possessed desirable properties, including negligible sive nanocarrier, indicating β-CD–capped UiO-68-azo was stimuli
premature release, non-cytotoxicity, and good biocompatibility. responsive and without premature release. This work provided a
In addition, β-cyclodextrin (β-CD) is also one of the most promising platform for controllable drug delivery.
promising supramolecular macrocycles that can be tethered onto Some other attempts have been made to design multirespon-
the scaffold MOFs as gating entitles to regulate targeted drug sive MOFs carriers in addition to the above-mentioned supra-
release. For example, the Zhang group introduced a multifunc- molecular nanovalves systems. For instance, two biocompatible
tional MOF platform for drug delivery by one-pot post-synthetic MOFs Fe-BTC and Zn-BTC (BTC = 1,3,5-benzenetricarbox-
surface modification of MIL-101 (Figure 8a).[48] After encapsu- ylic acid) have been investigated for one-pot encapsulation of
lating DOX, MIL-101-N3 (Fe) was coated with β-CD derivative DOX at room temperature.[50] Drug release could be triggered
(β-CD-SS-BCN) and a targeted peptide functionalized polymer, in the presence of biocompatible liposomes and lower pH, as
Lys (adamantane)-Arg-Gly-Asp-Ser-bi-poly(ethylene glycol) (PEG) carboxylate anions are protonated in acidic environments and
1900 (bi = benzoic imine bond, K(ad)RGDS-PEG1900), could be break down the coordination between carboxylic acid group and
tethered to the surface of the nanocarrier via a host–guest inter- metal center, leading to drug release by disruption of the MOF.
action. The proposed drug carrier possessed pH-responsiveness Furthermore, stronger electrostatic interactions between DOX
and redox-responsiveness due to the presence of a benzoic and liposomes also promoted the release of drugs. The obtained
imine bond in K(ad)RGDS-PEG1900 and disulfide bonds upon MOFs nanocarrier was initially used to release DOX in the pres-
linking β-CD on the MOF. Experimental results indicated that ence of liposomes and low-acidity conditions, notably affording
the suggested DDS possessed enhanced tumor cell uptake and new opportunities in stimuli-responsive drug vehicles.
that reducing agents triggered drug release, resulting in effi-
cient drug delivery and cancer therapy (Figure 8b). Moreover,
surface modification reduced the cytotoxicity of the loaded DOX 4. Multifunctional MOFs for Cancer Theranostics
on normal cells according to in vitro experiments, meanwhile in
vivo experiments proved high chemotherapy efficacy of the DDS. Although numerous MOF-based therapeutic platforms have
Recently, Wang and co-workers reported another dual been widely developed in recent years, there are still many chal-
stimuli-responsive MOF for on-command cargo release.[49] lenges that need to be addressed such as current treatment

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (11 of 20) 1606134
www.advmat.de www.advancedsciencenews.com
Review

Figure 8. a) Schematic fabrication process of the multifunctional DDS based on MIL-101 and b) its cancer therapy procedure. Reproduced with permis-
sion.[48] Copyright 2015, Royal Society of Chemistry.

Figure 9. a) Schematic illustration of the synthesis of dual-stimuli-responsive DDS based on UiO-68-azo MOF capped by β-CD. b) Schematic represen-
tation of light triggered release. c) Schematic representation of step-by-step release. Reproduced with permission under the terms of the CC-BY-NC-3.0
unported license.[49] Copyright 2014, The Royal Society of Chemistry.

1606134 (12 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

modality, which is still limited in traditional chemotherapy and with c(RGDfK) and rhodamine B.[84] Experimental results veri-

Review
whose therapeutic efficacy is hampered by multidrug resistance. fied the target-specific uptake of the c(RGDfK)-modified Mn
Considerable effort has been devoted to the fabrication of multi- MOF by angiogenic cancer cells and their potential for MRI.
functional theranostic nanomedical platforms, such as versatile In 2009, they reported another theranostic platform based on
MOF-based diagnostic and/or therapeutic systems, to overcome iron-carboxylate MOF MIL-101.[51] In this work, optical imaging
aforementioned drawbacks to achieving superior anti-tumor contrast agent, 1,3,5,7-tetramethyl-4,4- difluoro-8-bromome-
efficacy, among which core–shell MOFs nanocomposites have thyl-4-bora-3a,4a-diaza-s-indacene (Br-BODIPY), and ethoxy-
shown marvelous potential as nanomedical platforms. In this succinato-cisplatin (ESCP) prodrug, c,c,t-[PtCl2 (NH3)2 (OEt)
section, we will mainly introduce core–shell structures of MOF (O2CCH2CH2CO2H)], were post-synthetically modified on the
nanocarriers, including polymer-coated MOFs, magnetic core– MOF. Because of the instability of the MIL-101 (Fe) in PBS
shell MOFs, and other core–shell MOF-based nano­composites. buffer, a thin layer of silica was coated on the surface of NPs
Furthermore, some multifunctional therapeutic nanoplatforms to slow down cargo release. Silyl-derived c(RGDfK) was subse-
constructed from MOFs will also be described. quently functionalized onto the silica shell to enhance cellular
uptake. As expected, the multifunctional MOF possessed appre-
ciable imaging and anticancer efficacy.
4.1. Polymer-Coated MOFs for Cancer Theranostics Although the above-mentioned silica-coated MOFs possessed
high drug-loading capacity, such nanoplatforms were limited by
Since the Lin group reported the first example of surface func- a burst release of drugs. Thus, the Lin group developed a new
tionalization of a MOF material with a silica shell for the con- cisplatin-containing MOF composed of disuccinatocisplatin
trolled release of metal constituents and luminescence sensing (DSCP) and ZrCl4, which was further stabilized by a lipid layer
of dipicolinic acid,[82] an array of multifunctional core–shell composed of cholesterol (chol), 1,2-dioleoyl-sn-glycero-3-phos-
MOF nanocomposites have been developed for biomedical phocholine (DOPC) and 1,2-distearoyl-sn-glycero-3-phosphoe-
applications due to their unique advantages, including enhanced thanolamine-N-PEG (DSPE–PEG2 K).[85] A targeting ligand
stability, water dispersibility, biocompatibility, and tunable func- anisamide (AA) that displayed an affinity for sigma receptors
tionality. For instance, in 2008 the Lin group designed Pt-con- was functionalized on the surface of the nanocomposites to
taining nanoscale coordination polymers (NCPs) constructed enhance cellular uptake. This formulation proved good anti-
from Tb3+ ions and c,c,t-(diamminedichlorodisuccinato) Pt cancer efficacy and allowed for the preparation of other lipid-
(IV) (DSCP), followed by a silica coating on the surface of the coated NCPs for nanomedical applications.
as-synthesized NCPs to enhance their biocompatibility and Afterwards, they reported another Mn-bisphosphonate MOF
further functionality (Figure 10).[83] The controlled release coated with lipid for theranostic applications.[52] The Mn-bis-
behavior of silica-coated NCPs suggested that the nanocompos- phosphonate MOF was constructed from zoledronic acid and
ites could efficiently control the release of Pt species by varying MnCl2 in the presence of 1,2-dioleoyl-sn-glycero-3-phosphate
the thickness of the silica shell. Silyl-derived c(RGDfK) was sodium salt (DOPA), indicating high loadings of the zole-
then grafted on the surface of the pre-synthesized nanocompos- dronate and considerable MRI property. Accordingly, the MOF
ites to enhance cellular uptake. In vitro experiments exhibited was coated with a lipid/lipid–PEG layer to improve drug-release
high anticancer efficacies and good biocompatibility of the Pt- kinetics and biocompatibility, and further modified with a tar-
based NCPs. geting group (anisamide) for specific cancer cell uptake. Such a
Subsequently, they reported core–shell nanostructures of theranostic nanoplatform demonstrated physiological stability,
Mn MOF coated with a thin silica shell and functionalized biocompatibility and imaging-guided cancer-target specificity,
facilitating a huge potential application in the
field of theranostic MOFs materials.
Horcajada and co-workers introduced
an example of a specific modification of
nanoscale MIL-100(Fe) with heparin to
endow the MOF with appreciable bio-
logical properties via a one-pot method.[53]
Compared with unmodified MIL-100(Fe),
heparin-coated nanocomposites evidenced
their colloidal stability and preserved their
ability to accommodate high caffeine load-
ings with a more progressive release. More
importantly, the heparin-coated MOF showed
potential stealth properties that resulted from
the influence of the coating on the cell tox-
icity and uptake, the activation of the com-
plement pathway, the cytokines production,
Figure 10. Schematic fabrication process of the silica-coated MOF for drug delivery at spe- macrophage uptake, cytokine profile, and
cific site of cancer cells. Reproduced with permission.[83] Copyright 2008, American Chemical reactive oxygen species (ROS) production.
Society. Certainly, this surface modification method

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (13 of 20) 1606134
www.advmat.de www.advancedsciencenews.com

paved a new way to the design of desirable MOFs for thera- ZIF-90 drug carrier combined efficient diagnosis and chemo-
Review

nostic applications. therapy in one system because the magnetic core provided MRI
Additionally, Tendeloo and co-workers were able to fabri- and magnetic thermal response simultaneously. Significantly,
cate poly(ε-caprolactone) coated Zr-based MOFs for anticancer ZIF-90 was scaled down to 64 nm and modified with rat serum
drug carriers.[54] In this work, poly(ε-caprolactone)–tocopheryl albumin to improve their stability in a physiological environ-
poly(ethylene glycol)-succinate (PCL–TPGS) copolymer was ment and facilitated targeted delivery.
engineered on the surface of UiO-66 and UiO-67 MOFs due to The Sahu group also developed multifunctional magnetic
its biocompatibility and non-toxicity. Then, two model drugs, core–shell MOFs as theranostic nanoplatforms. They intro-
hydrophilic cisplatin and hydrophobic taxol, were used to inves- duced isoreticular MOF (IRMOF-3) coated Fe3O4 NPs for
tigate the drug-loading capacity and drug-release behavior of paclitaxel delivery, followed by modification with fluorescent
the drug-loaded nanocarriers. In vitro drug-release measure- molecule rhodamine B isothiocyanate and folic acid.[57] Their
ments indicated a sustained drug release and decreased burst research demonstrated that the integration of targeted drug
effects. Moreover, the developed polymer-coated MOFs showed delivery, MRI, cell imaging, and chemotherapy was realized
low toxicity as a result of the MTT assay using two cell lines in the multifunctional porous nanoplatform. Subsequently, a
(U-87 MG and HSC-3). All of these results suggested that such similar IRMOF-3 coated Fe3O4 nanocomposite was developed
polymer-coated MOFs formulations were promising for further as targeted anticancer nanoplatform in their work.[86] Spe-
biomedical applications. cially, a biodegradable O-carboxymethyl chitosan (OCMC) was
incorporated on the surface of a Fe3O4 core, instead of Fe3O4,
to improve loading capacity and pH sensitivity. More impor-
4.2. Magnetic Core–Shell MOFs for Cancer Theranostics tantly, carbon dots (CDs) were encapsulated in the magnetic
nanocomposites to realize optical imaging for the first time.
The employment of iron oxide as a core to fabricate MOF-based The hybrid drug carrier exhibited high dosage of 1.63 g DOX
theranostic probes for cancer diagnosis and therapy have drawn g−1 magnetic MOF with pH responsive drug release, magnetic
considerable attentions because of their superior magnetic guided drug delivery, cancer cell targeting, optical imaging, and
properties in, for example, MRI, magnetic hyperthermia, and MRI. Effective theranostic efficacy, biocompatibility, and facile
magnetic separation. For instance, Fu and co-workers reported functionality made the magnetic MOF formulation a promising
a theranostic Fe3O4@UiO-66 core–shell nanocomposite that candidate for biomedical applications.
simultaneously performed drug delivery and MRI in vitro and In 2015, Wang and co-workers reported multifunctional
in vivo.[55] The shell UiO-66 was used to encapsulate DOX and Fe3O4@poly(acrylic acid)/Au nanoclusters/zeolitic imida-
the strong superparamagnetic core Fe3O4 could be quickly col- zolate framework-8 (ZIF-8) NPs (Fe3O4@PAA/AuNCs/ZIF-8
lected under an external magnetic field and ensured its ability NPs) for cancer diagnosis and visualized-synergistic therapy
for MRI. (Figure 11a).[58] The obtained nanomedical system possessed
In addition, Ding and co-workers designed another Fe3O4 many therapeutic properties, such as facile magnetic separa-
core–shell MOF (Fe3O4@ZIF-90) for accurate and visible drug tion, high DOX loading capacity, tri-modal imaging, dual pH-
delivery.[56] Considering the deep penetration capacity of mag- responsive drug release, and high in vivo tumor inhibition effi-
netic field stimulus, an extremely low frequency alternating cacy (Figure 11b). Tri-modal imaging of magnetic resonance,
magnetic field (ELF-AMF) with a frequency of 20 Hz was X-ray computed tomography, and optical imaging was utilized
proved to be able to accelerate drug release from the magnetic to combine the superiorities and circumvent the limitations
ZIF-90 because of the magnetic thermal effect. The Fe3O4@ of different imaging techniques. Thus, such multifunctional

Figure 11. a) Schematic illustration of the preparation of Fe3O4@PAA/AuNCs/ZIF-8 composite NPs for simultaneous tri-modal cancer imaging and
chemotherapy. b) Photographs of excised tumors from euthanized mice in the three groups. Reproduced with permission.[58] Copyright 2015, Royal
Society of Chemistry.

1606134 (14 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

could produce reactive singlet oxygen (1O2),

Review
and both of the magnetic core clusters, Fe3O4
and Au25(Capt)18−, could provide hyperthemal
effects (Figure 13a). Additionally, Fe3O4 pre-
sented magnetic targeting and MRI imaging
capabilities (Figure 13b). More importantly,
in vivo control experiments verified that the
combined theranostic mechanisms (photo­
dynamic therapy/photothermal therapy/mag-
netism) suggested a higher inhibition effect
than that of photodynamic therapy and/or
photothermal therapy (Figure 13c), and the
multifunctional Fe3O4/ZIF-8-Au25 nanocom-
posites showed high tumor inhibition effi-
cacy without side effects on normal organs.
All of these results confirmed superior thera-
peutic efficacy of the obtained theranostic
nanoplatform, which combined a therapeutic
mechanism and diagnostic capability in one
system, making this system a good candidate
for biomedical applications.

4.3. Other Versatile Core–Shell MOFs


Nanocomposites for Cancer Theranostics

Great effort has been made to design multi­


functional core–shell MOFs as efficient
theranostic nanoplatforms, in addition to
above-mentioned magnetic core–shell MOFs
theranostic systems, and many other versatile
core–shell MOF nanocomposites have shown
Figure 12. a) Schematic presentation of magnetic-field-guided DHA-loaded FCM NPs entering considerable potential for nanomedical appli-
tumor cells. b) The anticancer mechanism of the DHA delivery system. Reproduced with per- cations.[87] For example, Wang and co-workers
mission.[59] Copyright 2016 Elsevier Ltd. introduced a versatile Mn3[Co(CN)6]2@
SiO2@Ag nanocube platform for synergistic
core–shell nanocarriers showed great potential for efficient cancer treatment.[61] The inner core of the Mn3[Co(CN)6]2 MOF
theranostic applications. structure served as a T1–T2 dual-modal MRI imaging agent and
Another interesting study for the multifunctional magnetic two-photon fluorescence (TPF) imaging agent; the outer shell of
core–shell MOF theranostic platform was reported by Chen and Ag NPs endowed the system with photothermal therapy capa-
co-workers.[59] Multifunctional Fe3O4@C@MIL-100(Fe) (FCM) bility and improved TPF imaging at the same time. DOX was
nanocomposites were designed for simultaneous delivery of loaded in the multifunctional nanoplatform with a high loading
dihydroartemisinin (DHA) and Fe (III) as cancer theranostic capacity of 600 mg g−1, and drug release could be triggered
nanoplatforms (Figure 12a). When Fe (III) reached the acidic by light due to surface plasmon resonance (SPR) of the noble
environment of tumor tissue, it would be reduced to Fe (II) and Ag NPs. Controlled experiments indicated that the combined
then reacted with pH-responsive released DHA to yield ROS therapeutic effect of chemotherapy and photothermal therapy
that induce cancer cell death. In addition, the magnetic core was superior to sole chemotherapy or photothermal therapy. All
of Fe3O4@C, with properties of magnetic guidance and MRI, of these results suggested that the Mn3[Co(CN)6]2@SiO2@Ag
was further used to encapsulate CDs for fluorescence optical core–shell nanocomposites would be promising candidates for
imaging (FOI) (Figure 12b). In vitro and in vivo experiments an imaging-guided cancer synergistic therapeutic platform.
exhibited dual-modal imaging-guided pH-responsive chemo- Subsequently, the Chen group fabricated core–shell PB@
therapy improved diagnostic and therapeutic efficacy for cancer MIL-100(Fe) dual MOF (d-MOFs) nanocomposites, integrating
treatment. diagnosis and therapy into a single system (Figure 14a,b).[62]
Interestingly, Yang et al. fabricated a cooperative thera- Multimodal imaging was introduced in the nanoplatform
peutic system based on a multifunctional magnetic core–shell because the d-MOFs possessed T1/T2 and T2 dual-modal MRI
MOF.[60] The obtained Fe3O4/ZIF-8-Au25 nanocomposites as well as FOI capabilities. More importantly, the inner Prus-
integrated photodynamic/photothermal therapy, magnetic tar- sian Blue NPs were used for photothermal therapy in the NIR
geting, and MRI imaging into one theranostic system. Photody- region. Furthermore, the as-prepared nanoplatform with high
namic effects were induced because the Au25(Capt)18− clusters artemisinin loading capacity of 848.4 mg g–1 was pH-responsive

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (15 of 20) 1606134
www.advmat.de www.advancedsciencenews.com
Review

Figure 13. a) Infrared thermal images of a tumor-bearing mouse after injection of Fe3O4/ZIF-8 Au25-5mL as a function of the irradiation time under
808 nm NIR light (0.5 W cm−2). b) MR images before (left) and after (right) injection of Fe3O4/ZIF-8 Au25 in vivo. c) Photographs of the tumor tissues
excised from tumor-bearing mice treated with normal saline, pure NIR laser, pure IZ, pure Au25(Capt)18− clusters with NIR-laser irradiation, IZA with or
without NIR-laser irradiation, and IZA with two types of stimuli (NIR-laser irradiation (L) and magnet remote control (M)) on the 14th day, as well as
corresponding digital photographs of representative mice. Reproduced with permission.[60] Copyright 2015, Royal Society of Chemistry.

due to the degradation of outer MOFs in an acidic patho- in vivo experiments verified that the Mn-IR825@PDA–PEG
logical environment. In vitro and in vivo experiments proved nanoplatform possessed good biocompatibility and high photo-
that the obtained core–shell PB@MIL-100(Fe) possessed high thermal therapy efficiency. Importantly, it could be degraded in
antitumor efficiency (Figure 14c,d) by combing multimodal vivo and showed rapid clearance via the renal excretion system.
imaging diagnosis, pH-responsive drug release, and chemo- The developed work demonstrated great potential of such ther-
photothermal therapy in a single system. Similarly, Zhang and apeutic formulation for cancer treatment in vivo.
co-workers reported another PB@mSiO2–PEG multifunctional Interestingly, they also reported a hafnium-based MOF built
nanoplatform with high DOX loading capacity.[88] In vitro and from Hf4+ and tetrakis (4-carboxyphenyl) porphyrin (TCPP).[64]
in vivo experiments showed that such formulation has excel- This formulation integrated photodynamic therapy and radia-
lent antitumor capability by integrating MRI, photoacoustic tion therapy in one system due to Hf4+, which could intrinsi-
imaging, pH-sensitive release, and photothermal-chemotherapy cally perform as a radio-sensitizer to improve radiation therapy,
in one system. and the organic building block of TCPP could serve as photo­
Recently, Liu and co-workers reported a core–shell MOF sensitizer for photodynamic therapy. Experimental results
for imaging-guided photothermal therapy; the first example of exhibited that the developed nanoplatform possessed desirable
using MOFs as a contrast agent to guide photothermal therapy biocompatibility owing to the outer coating of the PEG poly-
of cancer.[63] In their work, Mn-IR825 MOF was the core, which mers and appreciable antitumor efficacy due to the combina-
offered MRI and photothermal conversion properties in the tion of photodynamic and radiation therapy. Importantly, this
obtained nanoplatform, and the outer coating of polydopamine multifunctional MOF-based nanoplatform showed no obvious
(PDA) was used for facile modification of PEG to confer the side effects to normal organs and could be cleared away by the
nanocomposites desirable physiological stability. In vitro and metabolic system of the modal mouse.

1606134 (16 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

Review
Figure 14. a) Schematic illustrations of the loading and targeting delivery process of d-MOFs in tumors. b) pH-responsive degradation of outer MOFs
for drug release and dual-modal FOI- and MRI-guided cancer therapy in vitro and in vivo. c) Tumor growth curves of mice after controllable treatments.
d) Body weights of mice for varied time periods after controllable treatments. Reproduced with permission.[62] Copyright 2016 Elsevier Ltd.

In addition, developments of core–shell MOF nanocom- nanoplatform possessed high sensitivity, low toxicity, and good
posites for feasible, accurate, and efficient diagnosis have biocompatibility. Their work opened new avenues to imaging-
also attracted increasing attention. For instance, Yang and guided cancer therapies by integrating the advantages of both
co-workers reported a multifunctional MOF for dual-modal UCNPs and MOFs in a single system.
imaging of tumors in vitro and in vivo.[65] The inner core of the
MOF was constructed from 1,1′-dicarboxyl ferrocene (Fc) and
magnetic gadolinium (III) ions, and an outer shell of SiO2 was 4.4. Flexible MOF-Based Theranostic Nanoplatforms
used for further conjugation of fluorescent dye RBITC and tar-
geting ligand RGD, to provide their strong luminescence and Recent development of MOF preparation techniques have paved
targeting capabilities, respectively. The pre-synthesized multi- the way toward multitargeted nanomedical applications in both
functional Fc-Gd@SiO2(RBITC)–RGD nanocomposites, with diagnosis and therapy, due to the facile integration of multi-
properties of low cytotoxicity, good water-dispersion, and high functional components via inclusion and post-synthetic modifi-
stability, enabled dual-modal T1- and T2-weighted MRI in vivo, cations of MOFs, together with the mature synthesis of MOFs.
providing a versatile platform for targeting and imaging of In addition to the aforementioned strategies, drug molecules
diagnostic systems. as building blocks for MOFs are a straightforward approach to
The Tang group introduced another core–shell UCNP@ enable the development of future theranostic or multimodal
Fe-MIL-101_NH2@PEG@FAs for fluorescent/magnetic dual- NPs. For example, Dastidar and co-workers reported various
mode targeted imaging.[66] In the multifunctional nanocom- MnII MOFs by incorporating nonsteroidal anti-inflammatory
posites, rare-earth-doped upconversion NPs (UCNPs) were drugs (NSAIDs) and the organic linker 1,2-bis(4-pyridyl)ethylene
used as the core to present upconversion luminescence (UCL) (L) as part of the material for cell imaging and drug delivery.[67]
properties, and an outer shell of Fe-MIL-101_NH2 MOF was Both of the synthesized MOFs showed potential for cell imaging
used for MRI, followed by modifying with NH2-PEG-COOH to and drug delivery because the imaging agent L and the drug
improve biocompatibility and stability. Subsequently, folic acid molecules were part of the coordination framework. Moreover,
was modified on the surface of the as-synthesized nanocom- good biocompatibility and biodegradability as well as comparable
posites for targeted imaging. In vitro and in vivo experiments anti-inflammatory responses of the MOFs endowed such formu-
verified that the tumor targeted dual-mode UCL/MR imaging lations with extensive potentials for theranostic bioapplications.

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (17 of 20) 1606134
www.advmat.de www.advancedsciencenews.com

Recently, such formulations have also been studied as therapy and/or diagnosis. As outlined here, many remarkable
Review

therapeutic systems, in which carboxyl-functionalized diiodo- MOF-based platforms have been developed for novel nano-
substituted BODIPYs (I2-BDP) were incorporated into the medical applications, ranging from individual MOF systems,
UiO-66 MOF for photodynamic therapy via solvent-assisted to functionalized MOF formulations, to current collaborative
ligand exchange (SALE).[68] The obtained UiO-PDT nanocrys- systems. However, there are still many challenges that need to
tals possessed low cytotoxicity in the dark with enhanced cel- be addressed for these systems before such nanocomposites
lular uptake and photostability, together with high efficiency to can be applied clinically. Firstly, more effort should be made to
generate 1O2 for further photodynamic therapy of cancer treat- fabricate versatile nontoxic or low-toxicity and good biocompat-
ment. All of these results demonstrate interesting strategies of ible MOFs to achieve prolonged blood circulation and to ensure
designing functional MOF-based biomedical nanoplatforms. that the decomposition products can be processed through
the body’s metabolic system. Despite many excellent reports,
research on the toxicity of MOFs and their biodegradation is still
5. Demands of MOFs for Bioapplications in its infancy. Future work may be focused on the fabrication
of non-toxic MOF carriers by selecting endogenous building
Considering the demands of MOFs for bio- and related applica- blocks or functionalizing MOFs with bioactive substances to
tions, properties such as the toxicological compatibility, biodeg- reduce side effects. Additionally, the stability and degradation
radability, and stability of MOFs need to be addressed. Firstly, mechanism of MOF-based nanocarriers need systematic studies
MOF materials should be toxicologically compatible in terms of in vitro, and systematic in vivo investigations are essential to
bioapplications, therefore it is essential that the MOF building optimize the performance of MOFs before clinical applications.
blocks are non-toxic. Currently, Fe, Zn, Zr, Mn, Mg, and Cu are To emphasize an important point, it is not enough to investi-
frequently applied for the construction of MOFs due to their gate the therapeutic effect of nanocarriers and their effect on
appropriate toxicity determined by oral lethal dose 50 (LD50), normal organs; further efforts need to be devoted to analyzing
i.e., 30 g kg–1 for Fe, 350 µg kg–1 for Zn, 4.1 g kg–1 for Zr, the metabolic mechanisms and metabolic pathways of nanocar-
1.5 g kg–1 for Mn, 8.1 g kg–1 for Mg, and 25 g kg–1 for Cu, riers in vivo. Importantly, subsequent long-term monitoring of
exhibiting their acceptable toxcity for bioapplications.[9e] As for organism is also indispensable. Furthermore, current studies
exogenous organic building blocks, terephthalic acid (LD50 = of imaging and therapeutic applications are far from being
5 g kg–1), trimesic acid (LD50 = 8.4 g kg–1), 1-methylimidazole applied clinically, and a comprehensive understanding of the
(LD50 = 1.13 g kg–1), and 2-methylimidazole (LD50 = 1.4 g kg–1), mechanism of absorption–distribution–metabolism–excretion is
are generally available for bioapplications. Furthermore, some needed. Finally, future endeavors should focus on the fabrica-
endogenous MOFs based on endogenous organic linker have tion of multimodal MOF-based theranostic platforms combined
been obtained. Ideally, such formulations would reduce severe with different mechanisms, for achieving excellent efficacy in
side effects.[29,40] In addition, some bioactive coatings are also anticancer or other diseases. Although theranostic MOFs for
acceptable solutions; for example, the above-mentioned MIL- clinical applications are still a long-standing challenge in nano-
100(Fe) with heparin coating was endowed with enhanced bio- medical development, scientists have made significant progress.
logical properties such as lower immunoresponse, improved It is expected that promising developments to MOFs for clinical
stability, and reduced cell recognition. To date, in vitro toxico- applications will be made.
logical experiments have been conducted to assess the cytotox-
icity of MOF-based nanocarriers determined by cell viability on
various cell lines[89] Still, further efforts should be devoted to Acknowledgements
the evaluation of toxicity in vivo, not only to the observation of
body and organ weights, but also to investigate metabolic path- The authors thank the National Natural Science Foundation of China
ways of such nanocarriers in vivo.[7a,69a,90] (51673084, 51473061) and the JLU Cultivation Fund for the National
Science Fund for Distinguished Young Scholars for financial support.
For bioapplications, the biodegradability and stability of The authors would also like to thank the journal editors and the
MOFs are other important concerns that need to be empha- reviewers who raised valuable points that help to improve the quality of
sized. The decomposition of MOF nanocarriers at a desired the presentation of this review article.
region is recommended to avoid endogenous accumulation.
Much research on the stabilities of diverse MOF materials Received: November 14, 2016
under simulated physiological conditions have been reported, Revised: December 18, 2016
Published online:
for example, MIL-101 (Fe) possessed poor stability in phosphate
buffer, while MIL-100 (Fe) is stable in water but decomposed
after several days.[7a,51] Thus, the stability and degradation
mechanism of various MOF carriers in vitro and in vivo need
further systematic investigation. [1] K. Cho, X. Wang, S. Nie, Z. G. Chen, D. M. Shin, Clin. Cancer Res.
2008, 14, 1310.
[2] T. L. Doane, C. Burda, Chem. Soc. Rev. 2012, 41, 2885.
6. Conclusion [3] a) Q. He, J. Shi, F. Chen, M. Zhu, L. Zhang, Biomaterials 2010, 31,
3335; b) W. A. Jamal, K. Kostarelos. Acc. Chem. Res. 2011, 44, 1094;
In summary, we have reviewed recent significant progress made c) C. Deng, Y. Jiang, R. Cheng, F. Meng, Z. Zhong, Nano Today 2012,
in adapting MOFs for drug delivery, especially for cancerous 7, 467; d) S. Karthik, N. Puvvada, B. N. Kumar, S. Rajput, A. Pathak,

1606134 (18 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134
www.advancedsciencenews.com www.advmat.de

M. Mandal, N. D. Singh, ACS. Appl. Mater. Interfaces 2013, 5, [19] J. Park, Q. Jiang, D. Feng, L. Mao, H. C. Zhou, J. Am. Chem. Soc.

Review
5232; e) R. G. Mendes, A. Bachmatiuk, B. Büchner, G. Cuniberti, 2016, 138, 3518.
M. H. Rümmeli, J. Mater. Chem. B 2013, 1, 401; f) F. Fu, Y. Wu, [20] P. Horcajada, C. Serre, M. Vallet-Regí, M. Sebban, F. Taulelle,
J. Zhu, S. Wen, M. Shen, X. Shi, ACS. Appl. Mater. Interfaces 2014, G. Férey, Angew. Chem. 2006, 118, 6120.
6, 16416. [21] P. Horcajada, C. Serre, G. Maurin, N. A. Ramsahye, F. Balas,
[4] a) Y. H. Choe, R. B. Greenwald, J. McGuire, C. D. Conover, Adv. M. Vallet-Regí, M. Sebban, F. Taulelle, G. Férey, J. Am. Chem. Soc.
Drug Delivery Rev. 2003, 55, 217; b) R. Tong, J. Cheng, Angew. Chem. 2008, 130, 6774.
2008, 120, 4908. [22] C. Y. Sun, C. Qin, C. G. Wang, Z. M. Su, S. Wang, X. L. Wang,
[5] a) S. Tang, X. Huang, X. Chen, N. Zheng, Adv. Funct. Mater. 2010, G. S. Yang, K. Z. Shao, Y. Q. Lan, E. B. Wang, Adv. Mater. 2011, 23,
20, 2442; b) P. Yang, S. Gai, J. Lin, Chem. Soc. Rev. 2012, 41, 3679. 5629.
[6] M. Gaumet, A. Vargas, R. Gurny, F. Delie, Eur. J. Pharm. Biopharm. [23] S. Rojas, F. J. Carmona, C. R. Maldonado, P. Horcajada, T. Hidalgo,
2008, 69, 1. C. Serre, J. A. Navarro, E. Barea, Inorg. Chem. 2016, 55, 2650.
[7] a) P. Horcajada, T. Chalati, C. Serre, B. Gillet, C. Sebrie, T. Baati, [24] P. P. Bag, D. Wang, Z. Chen, R. Cao, Chem. Commun. 2016, 52,
J. F. Eubank, D. Heurtaux, P. Clayette, C. Kreuz, J. S. Chang, 3669.
Y. K. Hwang, V. Marsaud, P. N. Bories, L. Cynober, S. Gil, G. Ferey, [25] N. M. Kazemi, S. A. Shojaosadati, A. Morsali, Microporous
P. Couvreur, R. Gref, Nat. Mater. 2010, 9, 172; b) R. C. Huxford, Mesoporous Mater. 2014, 186, 73.
J. Della Rocca, W. Lin, Curr. Opin. Chem. Biol. 2010, 14, 262; [26] C. He, K. Lu, D. Liu, W. Lin, J. Am. Chem. Soc. 2014, 136, 5181.
c) S. Mura, J. Nicolas, P. Couvreur, Nat. Mater. 2013, 12, 991; [27] S. Tai, W. Zhang, J. Zhang, G. Luo, Y. Jia, M. Deng, Y. Ling, Micropo-
d) C. He, D. Liu, W. Lin, Chem. Rev. 2015, 115, 11079; e) N. Song, rous Mesoporous Mater. 2016, 220, 148.
Y. W. Yang, Chem. Soc. Rev. 2015, 44, 3474; f) A. A. Moghanjoughi, [28] a) F. Vermoortele, B. Bueken, G. L. Bars, B. V. de Voorde,
D. Khoshnevis, A. Zarrabi, Drug Delivery Transl. Res. 2016, 6, 333. M. Vandichel, K. Houthoofd, A. Vimont, M. Daturi, M. Waroquier,
[8] a) T. Zhang, C. F. Chan, J. Hao, G. L. Law, W. K. Wong, K. L. Wong, V. V. Speybroeck, C. Kirschhock, D. E. De Vos, J. Am. Chem. Soc.
RSC Adv. 2013, 3, 382; b) S. Purushotham, R. V. Ramanujan, 2013, 135, 11465; b) D. Cunha, M. Ben Yahia, S. Hall, S. R. Miller,
Acta. Biomater. 2010, 6, 502; c) P. Vijayaraghavan, C. H. Liu, H. Chevreau, E. Elkaïm, G. Maurin, P. Horcajada, C. Serre, Chem.
R. Vankayala, C. S. Chiang, K. C. Hwang, Adv. Mater. 2014, 26, 6689; Mater. 2013, 25, 2767.
d) Z. H. Sheng, M. B. Zheng, P. F. Zhao, H. L. Liu, D. Y. Gao, [29] H. Oh, T. Li, J. An, Chem.–Eur. J. 2015, 21, 17010.
P. Gong, G. H. Gao, Y. F. Ma, L. T. Cai, ACS Nano 2014, 8, 12310; [30] P. F. Gao, L. L. Zheng, L. J. Liang, X. X. Yang, Y. F. Li, C. Z. Huang,
e) G. F. Luo, W. H. Chen, Q. Lei, W. X. Qiu, Y. X. Liu, Y. J. Cheng, J. Mater. Chem. B 2013, 1, 3202.
X. Z. Zhang, Adv. Funct. Mater. 2016, 26, 4339. [31] H. Ren, L. Zhang, J. An, T. Wang, L. Li, X. Si, L. He, X. Wu, C. Wang,
[9] a) M. G. Marqués, T. Hidalgo, C. Serre, P. Horcajada, Coord. Chem. Z. Su, Chem. Commun. 2014, 50, 1000.
Rev. 2016, 307, 342; b) K. K. Gangu, S. Maddila, S. B. Mukkamala, [32] X. Zhu, J. Gu, Y. Wang, B. Li, Y. Li, W. Zhao, J. Shi, Chem. Commun.
S. B. Jonnalagadda, Inorg. Chim. Acta. 2016, 446, 61; c) Y. Sun, 2014, 50, 8779.
H. C. Zhou, Sci. Technol. Adv. Mater. 2015, 16, 054202; d) W. Cai, [33] T. Kundu, S. Mitra, P. Patra, A. Goswami, D. Diaz Diaz, R. Banerjee,
C. C. Chu, G. Liu, Y. X. Wang, Small 2015, 11, 4806; e) P. Horcajada, Chem.–Eur. J. 2014, 20, 10514.
R. Gref, T. Baati, P. K. Allan, G. Maurin, P. Couvreur, G. Ferey, [34] J. Zhuang, C. H. Kuo, L. Y. Chou, D. Y. Liu, E. Weerapana,
R. E. Morris, C. Serre, Chem. Rev. 2012, 112, 1232. C. K. Tsung, ACS Nano 2014, 8, 2812.
[10] a) R. J. Kuppler, D. J. Timmons, Q. R. Fang, J. R. Li, T. A. Makal, [35] H. Zheng, Y. Zhang, L. Liu, W. Wan, P. Guo, A. M. Nystrom, X. Zou,
M. D. Young, D. Yuan, D. Zhao, W. Zhuang, H. C. Zhou, Coord. J. Am. Chem. Soc. 2016, 138, 962.
Chem. Rev. 2009, 253, 3042; b) C. Wang, D. Liu, W. Lin, J. Am. [36] Y. Yang, Q. Hu, Q. Zhang, K. Jiang, W. Lin, Y. Yang, Y. Cui, G. Qian,
Chem. Soc. 2013, 135, 13222. Mol. Pharmaceutics 2016, 13, 2782.
[11] a) E. D. Bloch, W. L. Queen, R. Krishna, J. M. Zadrozny, [37] W. Lin, Q. Hu, K. Jiang, Y. Yang, Y. Yang, Y. Cui, G. Qian, J. Solid
C. M. Brown, J. R. Long. Science 2012, 335, 1606; b) N. Ding, H. Li, State Chem. 2016, 237, 307.
X. Feng, Q. Wang, S. Wang, L. Ma, J. Zhou, B. Wang, J. Am. Chem. [38] F. Ke, Y. P. Yuan, L. G. Qiu, Y. H. Shen, A. J. Xie, J. F. Zhu, X. Y. Tian,
Soc. 2016, 138, 10100. L. D. Zhang, J. Mater. Chem. 2011, 21, 3843.
[12] A. W. Peters, Z. Li, O. K. Farha, J. T. Hupp, ACS Appl. Mater. Inter- [39] Y. N. Wu, M. Zhou, S. Li, Z. Li, J. Li, B. Wu, G. Li, F. Li, X. Guan,
faces 2016, 8, 20675. Small 2014, 10, 2927.
[13] a) R. B. Lin, S. Y. Liu, J. W. Ye, X. Y. Li, J. P. Zhang, Adv. Sci. 2016, [40] J. An, S. J. Geib, N. L. Rosi, J. Am. Chem. Soc. 2009, 131, 8376.
3, 1500434; b) T. Wang, Y. Jia, Q. Chen, R. Feng, S. Tian, T. L. Hu, [41] Q. Hu, J. Yu, M. Liu, A. Liu, Z. Dou, Y. Yang, J. Med. Chem. 2014,
X. H. Bu, Sci. China Chem. 2016, 59, 959. 57, 5679.
[14] O. R. Evans, W. Lin, Acc. Chem. Res. 2002, 35, 511. [42] S. Nagata, K. Kokado, K. Sada, Chem. Commun. 2015, 51, 8614.
[15] a) A. C. McKinlay, R. E. Morris, P. Horcajada, G. Ferey, R. Gref, [43] W. X. Lin, Q. Hu, J. C. Yu, K. Jiang, Y. Y. Yang, S. C. Xiang, Y. J. Cui,
P. Couvreur, C. Serre, Angew. Chem., Int. Ed. Engl. 2010, 49, 6260; Y. Yang, Z. Y. Wang, G. D. Qian, ChemPlusChem 2016, 81, 804.
b) P. Horcajada, R. Gref, T. Baati, P. K. Allan, G. Maurin, [44] K. Jiang, L. Zhang, Q. Hu, D. Zhao, T. Xia, W. Lin, Y. Yang, Y. Cui,
P. Couvreur, G. Ferey, R. E. Morris, C. Serre, Chem. Rev. 2012, 112, Y. Yang, G. Qian, J. Mater. Chem. B 2016, 4, 6398.
1232; c) R. Liu, T. Yu, Z. Shi, Z. Wang, Int. J. Nanomed. 2016, 11, [45] L. L. Tan, H. Li, Y. C. Qiu, D. X. Chen, X. Wang, R. Y. Pan, Y. Wang,
1187. S. X. A. Zhang, B. Wang, Y. W. Yang, Chem. Sci. 2015, 6, 1640.
[16] a) P. Silva, S. M. Vilela, J. P. Tome, F. A. Almeida Paz, Chem. Soc. [46] L. L. Tan, H. Li, Y. Zhou, Y. Zhang, X. Feng, B. Wang, Y. W. Yang,
Rev. 2015, 44, 6774; b) S. Li, F. Huo, Nanoscale 2015, 7, 7482. Small 2015, 11, 3807.
[17] a) J. D. Rocca, W. Lin, Eur. J. Inorg. Chem. 2010, 2010, 3725; [47] L. L. Tan, N. Song, S. X. A. Zhang, H. Li, B. Wang, Y. W. Yang,
b) J. D. Rocca, D. M. Liu, W. B. Lin. Acc. Chem. Res. 2011, 44, 957; J. Mater. Chem. B 2016, 4, 135.
c) C. Y. Sun, C. Q, X. L. Wang, Z. M. Su, Expert Opin. Drug Deliv. [48] X. G. Wang, Z. Y. Dong, H. Cheng, S. S. Wan, W. H. Chen,
2013, 10, 89. M. Z. Zou, J. W. Huo, H. X. Deng, X. Z. Zhang, Nanoscale 2015, 7,
[18] a) W. Lin, W. J. Rieter, K. M. Taylor, Angew. Chem., Int. Ed. 2009, 16061.
48, 650; b) O. K. Farha, J. T. Hupp. Acc. Chem. Res. 2010, 43, 1166; [49] X. S. Meng, B. Gui, D. Q. Yuan, M. Zeller, C. Wang, Sci. Adv. 2016,
c) H. C. Zhou, J. R. Long, O. M. Yaghi, Chem. Rev. 2012, 112, 673. 2, e1600480.

Adv. Mater. 2017, 1606134 © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com (19 of 20) 1606134
www.advmat.de www.advancedsciencenews.com
Review

[50] C. Adhikari, A. Chakraborty, ChemPhysChem 2016, 17, L. Trahms, S. Odenbach, C. Alexiou, J. Phys.: Condens. Matter
1070. 2006, 18, S2893; c) C. S. Kumar, F. Mohammad, Adv. Drug Deliv.
[51] K. M. L. Taylor-Pashow, J. D. Rocca, Z. G. Xie, S. Tran, W. B. Lin, Rev. 2011, 63, 789; d) N. Lee, T. Hyeon, Chem. Soc. Rev. 2012, 41,
J. Am. Chem. Soc. 2009, 131, 14261. 2575.
[52] D. Liu, C. He, C. Poon, W. Lin, J. Mater. Chem. B 2014, 2, 8249. [75] M. W. Freeman, A. Arrott, J. H. L. Watson, J. Appl. Phys. 1960, 31,
[53] E. Bellido, T. Hidalgo, M. V. Lozano, M. Guillevic, R. Simon-Vazquez, 404.
M. J. Santander-Ortega, A. Gonzalez-Fernandez, C. Serre, [76] D. Roy, W. L. Brooks, B. S. Sumerlin, Chem. Soc. Rev. 2013, 42,
M. J. Alonso, P. Horcajada, Adv. Healthcare Mater. 2015, 4, 1246. 7214.
[54] M. Filippousi, S. Turner, K. Leus, P. I. Siafaka, E. D. Tseligka, [77] a) T. Ogoshi, S. Kanai, S. Fujinami, T. Yamagishi, and Y. Nakamoto,
M. Vandichel, S. G. Nanaki, I. S. Vizirianakis, D. N. Bikiaris, J. Am. Chem. Soc. 2008, 130, 5022; b) N. L. Strutt, H. Zhang,
P. V. D. Voort, G. V. Tendeloo, Int. J. Pharm. 2016, 509, 208. S. T. Schneebeli, J. F. Stoddart, Acc. Chem. Res. 2014, 47, 2631;
[55] H. X. Zhao, Q. Zou, S. K. Sun, C. Yu, X. Zhang, R. J. Li, Y. Y. Fu, c) H. Zhang, Y. Zhao, Chem. Eur. J. 2013, 19, 16862; d) W. Si, P. Xin,
Chem. Sci. 2016, 7, 5294. Z. T. Li, J. L. Hou, Acc. Chem. Res. 2015, 48, 1612.
[56] J. Fang, Y. Yang, W. Xiao, B. Zheng, Y. B. Lv, X. L. Liu, J. Ding, [78] M. Xue, X. D. Chi, Z. B. Zhang, F. H. Huang, Acc. Chem. Res. 2012,
Nanoscale 2016, 8, 3259. 45, 1294.
[57] A. Ray Chowdhuri, D. Bhattacharya, S. K. Sahu, Dalton Trans. 2016, [79] a) Y. Wang, G. Ping, C. Li, Chem. Commun. 2016, 52, 9858;
45, 2963. b) Y. W. Yang, Y. L. Sun, N. Song, Acc. Chem. Res. 2014, 47, 1950;
[58] R. Bian, T. Wang, L. Zhang, L. Li, C. Wang, Biomater. Sci. 2015, 3, c) L. L. Tan, H. Li, Y. Tao, S. X. Zhang, B. Wang, Y. W. Yang, Adv.
1270. Mater. 2014, 26, 7027.
[59] D. Wang, J. Zhou, R. Chen, R. Shi, G. Xia, S. Zhou, Z. Liu, N. Zhang, [80] H. Li, D. X. Chen, Y. L. Sun, Y. B. Zheng, L. L. Tan, P. S. Weiss,
H. Wang, Z. Guo, Q. Chen, Biomaterials 2016, 107, 88. Y. W. Yang, J. Am. Chem. Soc. 2013, 135, 1570.
[60] D. Yang, G. Yang, S. Gai, F. He, G. An, Y. Dai, R. Lv, P. Yang, [81] E. Aznar, M. Oroval, L. Pascual, J. R. Murguia, R. M. Manez,
Nanoscale 2015, 7, 19568. F. Sancenon, Chem. Rev. 2016, 116, 561.
[61] D. Wang, Z. Guo, J. Zhou, J. Chen, G. Zhao, R. Chen, M. He, Z. Liu, [82] W. J. Rieter, K. M. L. Taylor, W. B. Lin, J. Am. Chem. Soc. 2007, 129,
H. Wang, Q. Chen, Small 2015, 11, 5956. 9852.
[62] D. Wang, J. Zhou, R. Chen, R. Shi, G. Zhao, G. Xia, R. Li, Z. Liu, [83] W. J. Rieter, K. M. Pott, K. M. L. Taylor, W. B. Lin, J. Am. Chem. Soc.
J. Tian, H. Wang, Z. Guo, H. Wang, Q. Chen, Biomaterials 2016, 2008, 130, 11584.
100, 27. [84] W. J. Rieter, K. M. L. Taylor, W. B. Lin, J. Am. Chem. Soc. 2008, 130,
[63] Y. Yang, J. Liu, C. Liang, L. Feng, T. Fu, Z. Dong, Y. Chao, Y. Li, 14358.
G. Lu, M. Chen, Z. Liu, ACS Nano 2016, 10, 2774. [85] R. C. Huxford-Phillips, S. R. Russell, D. Liu, W. Lin, RSC Adv. 2013,
[64] J. Liu, Y. Yang, W. Zhu, X. Yi, Z. Dong, X. Xu, M. Chen, K. Yang, 3, 14438.
G. Lu, L. Jiang, Z. Liu, Biomaterials 2016, 97, 1. [86] A. R. Chowdhuri, T. Singh, S. K. Ghosh, S. K. Sahu, ACS Appl. Mater.
[65] H. Yang, C. Qin, C. Yu, Y. Lu, H. Zhang, F. Xue, D. Wu, Z. Zhou, Interfaces 2016, 8, 16573.
S. Yang, Adv. Funct. Mater. 2014, 24, 1738. [87] a) A. R. Chowdhuri, D. Laha, S. Pal, P. Karmakar, S. K. Sahu,
[66] Y. Li, J. Tang, L. He, Y. Liu, Y. Liu, C. Chen, Z. Tang, Adv. Mater. Dalton Trans. 2016, 45, 18120; b) W. Q. Wang, L. Wang, Y. Li, S. Liu,
2015, 27, 4075. Z. G. Xie, X. B. Jing, Adv. Mater. 2016, 28, 9320; c) M. Nazari,
[67] M. Paul, P. Dastidar, Chem. Eur. J. 2016, 22, 988. M. Rubio-Martinez, G. Tobias, J. P. Barrio, R. Babarao, F. Nazari,
[68] W. Wang, L. Wang, Z. Li, Z. Xie, Chem. Commun. 2016, 52, 5402. K. Konstas, B. W. Muir, S. F. Collins, A. J. Hill, M. C. Duke,
[69] a) T. Baati, L. Njim, F. Neffati, A. Kerkeni, M. Bouttemi, R. Gref, M. R. Hill, Adv. Funct. Mater. 2016, 26, 3244; d) X. P. Duan, C. Chan,
M. F. Najjar, A. Zakhama, P. Couvreur, C. Serre, P. Horcajada, N. N. Guo, W. B. Han, R. R. Weichselbaum, W. B. Lin, J. Am. Chem.
Chem. Sci. 2013, 4, 1597; b) H. Chevreau, A. Permyakova, F. Nouar, Soc. 2016, 138, 16686.
P. Fabry, C. Livage, F. Ragon, A. Garcia-Marquez, T. Devic, [88] Y. Y. Su, Z. Teng, H. Yao, S. J. Wang, Y. Tian, Y. L. Zhang, W. F. Liu,
N. Steunou, C. Serre, P. Horcajada, CrystEngComm 2016, 18, 4094. W. Tian, L. J. Zheng, N. Lu, Q. Q. Ni, X. D. Su, Y. X. Tang, J. Sun,
[70] J. H. Cavka, S. Jakobsen, U. Olsbye, N. Guillou, C. Lamberti, Y. Liu, J. Wu, G. F. Yang, G. M. Lu, L. J. Zhang, ACS Appl. Mater.
S. Bordiga, K. P. Lillerud, J. Am. Chem. Soc. 2008, 130, 13850. Interfaces 2016, 8, 17038.
[71] Y. Bai, Y. Dou, L. H. Xie, W. Rutledge, J. R. Li, H. C. Zhou, Chem. [89] a) D. M. Liu, S. A. Kramer, R. C. Huxford-Phillips, S. Z. Wang,
Soc. Rev. 2016, 45, 2327. J. D. Rocca, W. B. Lin, Chem. Commun. 2012, 48, 2668;
[72] Y. Lu, A. A. Aimetti, R. Langer, Z. Gu, Nat. Rev. Mater. 2016, 1, b) R. C. Huxford, K. E. Dekrafft, W. S. Boyle, D. M. Liu, W. B. Lin,
16075. Chem. Sci. 2012, 3, 198; c) A. Ruyra, A. Yazdi, J. Espin, A. Carne-
[73] S. Angelos, N. M. Khashab, Y. W. Yang, A. Trabolsi, H. A. Khatib, Sanchez, N. Roher, J. Lorenzo, I. Imaz, D. Maspoch, Chemistry
J. F. Stoddart, J. I. Zink, J. Am. Chem. Soc. 2009, 27, 7692. 2015, 21, 2508.
[74] a) R. Hergt, S. Dutz, R. Müller, M. Zeisberger, J. Phys.: Condens. [90] C. Tamames-Tabar, D. Cunha, E. Imbuluzqueta, F. Ragon, C. Serre,
Matter 2006, 18, S2919; b) R. Jurgons, C. Seliger, A. Hilpert, M. J. Blanco-Prieto, P. Horcajada, J. Mater. Chem. B 2014, 2, 262.

1606134 (20 of 20) wileyonlinelibrary.com © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2017, 1606134

You might also like