Please wait a minute...
Frontiers of Materials Science

ISSN 2095-025X

ISSN 2095-0268(Online)

CN 11-5985/TB

Postal Subscription Code 80-974

2018 Impact Factor: 1.701

Front. Mater. Sci.    2021, Vol. 15 Issue (3) : 374-390    https://doi.org/10.1007/s11706-021-0568-2
REVIEW ARTICLE
Metal-organic framework-based intelligent drug delivery systems for cancer theranostic: A review
Qingni XU, Chaohua LI, Yuqi CHEN, Yueli ZHANG, Bo LU()
School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
 Download: PDF(6124 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

The design and development of multifunctional nano-drug delivery systems (NDDSs) is a solution that is expected to solve some intractable problems in traditional cancer treatment. In particular, metal-organic frameworks (MOFs) are novel hybrid porous nanomaterials which are constructed by the coordination of metal cations or clusters and organic bridging ligands. Benefiting from their intrinsic superior properties, MOFs have captivated intensive attentions in drug release and cancer theranostic. Based on what has been achieved about MOF-based DDSs in recent years, this review introduces different stimuli-responsive mechanisms of them and their applications in cancer diagnosis and treatment systematically. Moreover, the existing challenges and future opportunities in this field are summarized. By realizing industrial production and paying attention to biosafety, their clinical applications will be enriched.

Keywords metal-organic framework      nanomaterial      stimuli-responsiveness      cancer theranostic     
Corresponding Author(s): Bo LU   
Online First Date: 07 September 2021    Issue Date: 24 September 2021
 Cite this article:   
Qingni XU,Chaohua LI,Yuqi CHEN, et al. Metal-organic framework-based intelligent drug delivery systems for cancer theranostic: A review[J]. Front. Mater. Sci., 2021, 15(3): 374-390.
 URL:  
https://academic.hep.com.cn/foms/EN/10.1007/s11706-021-0568-2
https://academic.hep.com.cn/foms/EN/Y2021/V15/I3/374
Fig.1  Schematic illustration for the formation and drug release mechanism of the HA/α-TOS@ZIF-8 nanoplatform. Reproduced with permission from Ref. [42].
Fig.2  Schematic illustration of the preparation of CCM@MOF-M(DTBA), and the redox-responsive degradation of CCM@MOF-M(DTBA) in tumor cells for cancer therapy. Reproduced with permission from Ref. [64].
Fig.3  Schematic illustration of SQU@PCN preparation and the tumor cell death process by homeostatic perturbation therapy and sensitized photodynamic therapy. Reproduced with permission from Ref. [70].
Fig.4  Schematic illustration for the formation and drug release mechanism of UCNPs@MIL-100(Fe) NPs. Reproduced with permission from Ref. [80].
Fig.5  (a) Scheme of the synthesis process for DHMS. (b) The proposed mechanism of ROS generation by DHMS under ultrasound irradiation. Reproduced with permission from Ref. [88].
Fig.6  Schematic illustration of the design and application of novel porous Fe3O4@C–PVP@DOX nanocomposites as smart platforms for the combined cancer therapy. Reproduced with permission from Ref. [91].
Fig.7  Schematic illustration of the preparation of NMIL-100@GOx@C and the cascade processes for cancer therapy. Reproduced with permission from Ref. [106].
Fig.8  (a) Synthesis of MnOx/UiO-66-F/PPEG NPs and 1H/19F dual-mode imaging of MnOx/UiO-66-F/PPEG NPs in vivo. (b) The representative 1H MRI, 19F MRI, and the merge of 1H MRI and 19F MRI under a 7 T magnetic field at different time (0, 30 and 90 min, respectively) in tumor and subcutaneous tissue with a dose of 249.4 mg·kg−1 body weight. Reproduced with permission from Ref. [118].
Fig.9  (a) Schematic illustration of the design and application of DOX/Pd@ZIF-8@PDA NPs. (b) PA imaging of DOX/Pd@ZIF-8@PDA NPs in tumor site. Reproduced with permission from Ref. [120].
Fig.10  (a) Schematic illustration of an endocytosis Mn3+-sealed MOF nanosystem for MRI- and OI-guided PDT by controlled ROS generation and GSH depletion after being unlocked by overexpressed GSH in tumor cells. (b) Fluorescence and (c) T1 contrast signals in tumor sites by intratumoral injection within 30 min. (d) In vivo MRI signal after intravenous injection with MOFs. (e) FI of mice over time by intravenous injection and tissue imaging at 36 h postinjection [122]. (f) Schematic illustration of the fabrication process of FDGI NPs. (g) Schematic illustration of FDGI theranostic nanoplatform for MR/PA/PT imaging-guided chemotherapy, PTT and PDT compound antitumor therapy [123]. Reproduced with permission from Refs. [122123].
1 H He, H Xie, Y Chen, et al.. Global, regional, and national burdens of bladder cancer in 2017: Estimates from the 2017 global burden of disease study. BMC Public Health, 2020, 20(1): 1693
https://doi.org/10.1186/s12889-020-09835-7 pmid: 33176751
2 H Sung, J Ferlay, R L Siegel, et al.. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians, 2021, 71(3): 209–249
https://doi.org/10.3322/caac.21660 pmid: 33538338
3 M Zhang, Y Ma, Z Wang, et al.. A CD44-targeting programmable drug delivery system for enhancing and sensitizing chemotherapy to drug-resistant cancer. ACS Applied Materials & Interfaces, 2019, 11(6): 5851–5861
https://doi.org/10.1021/acsami.8b19798 pmid: 30648841
4 A Kumar, V Jaitak. Natural products as multidrug resistance modulators in cancer. European Journal of Medicinal Chemistry, 2019, 176: 268–291
https://doi.org/10.1016/j.ejmech.2019.05.027 pmid: 31103904
5 J D Obayemi, A A Salifu, S C Eluu, et al.. LHRH-conjugated drugs as targeted therapeutic agents for the specific targeting and localized treatment of triple negative breast cancer. Scientific Reports, 2020, 10(1): 8212
https://doi.org/10.1038/s41598-020-64979-1 pmid: 32427904
6 I Mittra, K Pal, N Pancholi, et al.. Prevention of chemotherapy toxicity by agents that neutralize or degrade cell-free chromatin. Annals of Oncology, 2017, 28(9): 2119–2127
https://doi.org/10.1093/annonc/mdx318 pmid: 28911066
7 Y Ding, Y Ma, C Du, et al.. NO-releasing polypeptide nanocomposites reverse cancer multidrug resistance via triple therapies. Acta Biomaterialia, 2021, 123: 335–345
https://doi.org/10.1016/j.actbio.2021.01.015 pmid: 33476826
8 P Mi. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics, 2020, 10(10): 4557–4588
https://doi.org/10.7150/thno.38069 pmid: 32292515
9 S Raj, S Khurana, R Choudhari, et al.. Specific targeting cancer cells with nanoparticles and drug delivery in cancer therapy. Seminars in Cancer Biology, 2021, 69: 166–177
https://doi.org/10.1016/j.semcancer.2019.11.002 pmid: 31715247
10 H Liu, W Jiang, Q Wang, et al.. ROS-sensitive biomimetic nanocarriers modulate tumor hypoxia for synergistic photodynamic chemotherapy. Biomaterials Science, 2019, 7(9): 3706–3716
https://doi.org/10.1039/C9BM00634F pmid: 31187794
11 A Pandey, S Kulkarni, A P Vincent, et al.. Hyaluronic acid-drug conjugate modified core–shell MOFs as pH responsive nanoplatform for multimodal therapy of glioblastoma. International Journal of Pharmaceutics, 2020, 588: 119735
https://doi.org/10.1016/j.ijpharm.2020.119735 pmid: 32763386
12 V Sanfilippo, V C L Caruso, L M Cucci, et al.. Hyaluronan-metal gold nanoparticle hybrids for targeted tumor cell therapy. International Journal of Molecular Sciences, 2020, 21(9): 3085–3111
https://doi.org/10.3390/ijms21093085 pmid: 32349323
13 J Chen, Y Ma, W Du, et al.. Furin-instructed intracellular gold nanoparticle aggregation for tumor photothermal therapy. Advanced Functional Materials, 2020, 30(50): 2001566
https://doi.org/10.1002/adfm.202001566
14 M Kong, Y Huang, R Yu, et al.. Coordination bonding-based Fe3O4@PDA-Zn2+-doxorubicin nanoparticles for tumor chemo-photothermal therapy. Journal of Drug Delivery Science and Technology, 2019, 51: 185–193
https://doi.org/10.1016/j.jddst.2019.02.030
15 W Wen, L Wu, Y Chen, et al.. Ultra-small Fe3O4 nanoparticles for nuclei targeting drug delivery and photothermal therapy. Journal of Drug Delivery Science and Technology, 2020, 58: 101782
https://doi.org/10.1016/j.jddst.2020.101782
16 Y Guan, Y Yang, X Wang, et al.. Multifunctional Fe3O4@SiO2-CDs magnetic fluorescent nanoparticles as effective carrier of gambogic acid for inhibiting VX2 tumor cells. Journal of Molecular Liquids, 2021, 327: 114783
https://doi.org/10.1016/j.molliq.2020.114783
17 L Huang, J Liu, F Gao, et al.. A dual-responsive, hyaluronic acid targeted drug delivery system based on hollow mesoporous silica nanoparticles for cancer therapy. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2018, 6(28): 4618–4629
https://doi.org/10.1039/C8TB00989A pmid: 32254406
18 M Shao, C Chang, Z Liu, et al.. Polydopamine coated hollow mesoporous silica nanoparticles as pH-sensitive nanocarriers for overcoming multidrug resistance. Colloids and Surfaces B: Biointerfaces, 2019, 183: 110427
https://doi.org/10.1016/j.colsurfb.2019.110427 pmid: 31408782
19 K Chen, C Chang, Z Liu, et al.. Hyaluronic acid targeted and pH-responsive nanocarriers based on hollow mesoporous silica nanoparticles for chemo-photodynamic combination therapy. Colloids and Surfaces B: Biointerfaces, 2020, 194: 111166
https://doi.org/10.1016/j.colsurfb.2020.111166 pmid: 32521461
20 J Saleem, L Wang, C Chen. Carbon-based nanomaterials for cancer therapy via targeting tumor microenvironment. Advanced Healthcare Materials, 2018, 7(20): 1800525
https://doi.org/10.1002/adhm.201800525 pmid: 30073803
21 K P Loh, D Ho, G N C Chiu, et al.. Clinical applications of carbon nanomaterials in diagnostics and therapy. Advanced Materials, 2018, 30(47): 1802368
https://doi.org/10.1002/adma.201802368 pmid: 30133035
22 B P Jiang, B Zhou, Z Lin, et al.. Recent advances in carbon nanomaterials for cancer phototherapy. Chemistry, 2019, 25(16): 3993–4004
https://doi.org/10.1002/chem.201804383 pmid: 30328167
23 S Zhao, W Cao, S Xing, et al.. Enhancing effects of theanine liposomes as chemotherapeutic agents for tumor therapy. ACS Biomaterials Science & Engineering, 2019, 5(7): 3373–3379
https://doi.org/10.1021/acsbiomaterials.9b00317 pmid: 33405579
24 K Zhang, Y Zhang, X Meng, et al.. Light-triggered theranostic liposomes for tumor diagnosis and combined photodynamic and hypoxia-activated prodrug therapy. Biomaterials, 2018, 185: 301–309
https://doi.org/10.1016/j.biomaterials.2018.09.033 pmid: 30265899
25 Y Yang, X Liu, W Ma, et al.. Light-activatable liposomes for repetitive on-demand drug release and immunopotentiation in hypoxic tumor therapy. Biomaterials, 2021, 265: 120456
https://doi.org/10.1016/j.biomaterials.2020.120456 pmid: 33099066
26 L Yang, C Zhang, J Liu, et al.. ICG-conjugated and 125I-labeled polymeric micelles with high biosafety for multimodality imaging-guided photothermal therapy of tumors. Advanced Healthcare Materials, 2020, 9(5): 1901616
https://doi.org/10.1002/adhm.201901616 pmid: 31990442
27 C Hu, W Zhuang, T Yu, et al.. Multi-stimuli responsive polymeric prodrug micelles for combined chemotherapy and photodynamic therapy. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2020, 8(24): 5267–5279
https://doi.org/10.1039/D0TB00539H pmid: 32441291
28 S Zhong, C Chen, G Yang, et al.. Acid-triggered nanoexpansion polymeric micelles for enhanced photodynamic therapy. ACS Applied Materials & Interfaces, 2019, 11(37): 33697–33705
https://doi.org/10.1021/acsami.9b12620 pmid: 31487149
29 X Li, Y Zhang, X Zhi, et al.. Analysis of clinical efficacy of nano-albumin paclitaxel treatment for advanced cell lung cancer. Journal of Nanoscience and Nanotechnology, 2020, 20(10): 6019–6025
https://doi.org/10.1166/jnn.2020.18556 pmid: 32384947
30 Y Zhou, C Chang, Z Liu, et al.. Hyaluronic acid-functionalized hollow mesoporous silica nanoparticles as pH-sensitive nanocarriers for cancer chemo-photodynamic therapy. Langmuir, 2021, 37(8): 2619–2628
https://doi.org/10.1021/acs.langmuir.0c03250 pmid: 33586432
31 L Zhang, X Shi, Z Zhang, et al.. Porphyrinic zirconium metal-organic frameworks (MOFs) as heterogeneous photocatalysts for PET-RAFT polymerization and stereolithography. Angewandte Chemie International Edition, 2021, 60(10): 5489–5496
https://doi.org/10.1002/anie.202014208 pmid: 33179352
32 E Gulcay, I Erucar. Biocompatible MOFs for storage and separation of O2: A molecular simulation study. Industrial & Engineering Chemistry Research, 2019, 58(8): 3225–3237
https://doi.org/10.1021/acs.iecr.8b04084
33 H Daglar, H C Gulbalkan, G Avci, et al.. Effect of metal-organic framework (MOF) database selection on the assessment of gas storage and separation potentials of MOFs. Angewandte Chemie International Edition, 2021, 60(14): 7828–7837
https://doi.org/10.1002/anie.202015250 pmid: 33443312
34 M Hao, M Qiu, H Yang, et al.. Recent advances on preparation and environmental applications of MOF-derived carbons in catalysis. Science of the Total Environment, 2021, 760: 143333
https://doi.org/10.1016/j.scitotenv.2020.143333 pmid: 33190884
35 M J Neufeld, H Winter, M R Landry, et al.. Lanthanide metal-organic frameworks for multispectral radioluminescent imaging. ACS Applied Materials & Interfaces, 2020, 12(24): 26943–26954
https://doi.org/10.1021/acsami.0c06010 pmid: 32442367
36 X Gao, R Cui, G Ji, et al.. Size and surface controllable metal-organic frameworks (MOFs) for fluorescence imaging and cancer therapy. Nanoscale, 2018, 10(13): 6205–6211
https://doi.org/10.1039/C7NR08892B pmid: 29560986
37 P Kumar, B Anand, Y F Tsang, et al.. Regeneration, degradation, and toxicity effect of MOFs: Opportunities and challenges. Environmental Research, 2019, 176: 108488
https://doi.org/10.1016/j.envres.2019.05.019 pmid: 31295665
38 J Schnabel, R Ettlinger, H Bunzen. Zn-MOF-74 as pH-responsive drug-delivery system of arsenic trioxide. Chem-NanoMat, 2020, 6(8): 1229–1236
https://doi.org/10.1002/cnma.202000221
39 Z Liu, T Li, F Han, et al.. A cascade-reaction enabled synergistic cancer starvation/ROS-mediated/chemo-therapy with an enzyme modified Fe-based MOF. Biomaterials Science, 2019, 7(9): 3683–3692
https://doi.org/10.1039/C9BM00641A pmid: 31361291
40 Y Xiao, M Xu, N Lv, et al.. Dual stimuli-responsive metal-organic framework-based nanosystem for synergistic photothermal/pharmacological antibacterial therapy. Acta Biomaterialia, 2021, 122: 291–305
https://doi.org/10.1016/j.actbio.2020.12.045 pmid: 33359766
41 S Liang, X Xiao, L Bai, et al.. Conferring Ti-based MOFs with defects for enhanced sonodynamic cancer therapy. Advanced Materials, 2021, 33(18): 2100333
https://doi.org/10.1002/adma.202100333 pmid: 33792083
42 Q Sun, H Bi, Z Wang, et al.. Hyaluronic acid-targeted and pH-responsive drug delivery system based on metal-organic frameworks for efficient antitumor therapy. Biomaterials, 2019, 223: 119473
https://doi.org/10.1016/j.biomaterials.2019.119473 pmid: 31499255
43 Z Jiang, T Wang, S Yuan, et al.. A tumor-sensitive biological metal-organic complex for drug delivery and cancer therapy. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2020, 8(32): 7189–7196
https://doi.org/10.1039/D0TB00599A pmid: 32618980
44 W Pan, M Shi, Y Li, et al.. A GSH-responsive nanophotosensitizer for efficient photodynamic therapy. RSC Advances, 2018, 8(74): 42374–42379
https://doi.org/10.1039/C8RA08549H
45 L Y Yu, Y A Shen, M H Chen, et al.. The feasibility of ROS- and GSH-responsive micelles for treating tumor-initiating and metastatic cancer stem cells. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2019, 7(19): 3109–3118
https://doi.org/10.1039/C8TB02958J
46 E Sameiyan, E Bagheri, S Dehghani, et al.. Aptamer-based ATP-responsive delivery systems for cancer diagnosis and treatment. Acta Biomaterialia, 2021, 123: 110–122
https://doi.org/10.1016/j.actbio.2020.12.057 pmid: 33453405
47 L Yuwen, Q Qiu, W Xiu, et al.. Hyaluronidase-responsive phototheranostic nanoagents for fluorescence imaging and photothermal/photodynamic therapy of methicillin-resistant Staphylococcus aureus infections. Biomaterials Science, 2021, 9(12): 4484–4495
https://doi.org/10.1039/D1BM00406A pmid: 34002742
48 N Zhang, M Li, X Sun, et al.. NIR-responsive cancer cytomembrane-cloaked carrier-free nanosystems for highly efficient and self-targeted tumor drug delivery. Biomaterials, 2018, 159: 25–36
https://doi.org/10.1016/j.biomaterials.2018.01.007 pmid: 29309991
49 Y Ding, C Du, J Qian, et al.. NIR-responsive polypeptide nanocomposite generates NO gas, mild photothermia, and chemotherapy to reverse multidrug-resistant cancer. Nano Letters, 2019, 19(7): 4362–4370
https://doi.org/10.1021/acs.nanolett.9b00975 pmid: 31199153
50 W Zhang, J Lu, X Gao, et al.. Enhanced photodynamic therapy by reduced levels of intracellular glutathione obtained by employing a nano-MOF with Cu(II) as the active center. Angewandte Chemie International Edition, 2018, 57(18): 4891–4896
https://doi.org/10.1002/anie.201710800 pmid: 29451722
51 M Sabz, R Kamali, S Ahmadizade. Numerical simulation of magnetic drug targeting to a tumor in the simplified model of the human lung. Computer Methods and Programs in Biomedicine, 2019, 172: 11–24
https://doi.org/10.1016/j.cmpb.2019.02.001 pmid: 30902122
52 S Shen, D Huang, J Cao, et al.. Magnetic liposomes for light-sensitive drug delivery and combined photothermal-chemotherapy of tumors. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2019, 7(7): 1096–1106
https://doi.org/10.1039/C8TB02684J pmid: 32254777
53 T G Nguyen Cao, J H Kang, J Y You, et al.. Safe and targeted sonodynamic cancer therapy using biocompatible exosome-based nanosonosensitizers. ACS Applied Materials & Interfaces, 2021, 13(22): 25575–25588
https://doi.org/10.1021/acsami.0c22883 pmid: 34033477
54 M Mathesh, J Sun, F van der Sandt, et al.. Supramolecular nanomotors with “pH taxis” for active drug delivery in the tumor microenvironment. Nanoscale, 2020, 12(44): 22495–22501
https://doi.org/10.1039/D0NR04415F pmid: 33169767
55 T Yan, S Zhu, W Hui, et al.. Chitosan based pH-responsive polymeric prodrug vector for enhanced tumor targeted co-delivery of doxorubicin and siRNA. Carbohydrate Polymers, 2020, 250: 116781
https://doi.org/10.1016/j.carbpol.2020.116781 pmid: 33049806
56 I A Lázaro, S Haddad, S Sacca, et al.. Selective surface PEGylation of UiO-66 nanoparticles for enhanced stability, cell uptake, and pH-responsive drug delivery. Chem, 2017, 2(4): 561–578
https://doi.org/10.1016/j.chempr.2017.02.005 pmid: 28516168
57 K Jiang, L Zhang, Q Hu, et al.. Indocyanine green-encapsulated nanoscale metal-organic frameworks for highly effective chemo-photothermal combination cancer therapy. Materials Today Nano, 2018, 2: 50–57
https://doi.org/10.1016/j.mtnano.2018.09.001
58 S Yu, S Wang, Z Xie, et al.. Hyaluronic acid coating on the surface of curcumin-loaded ZIF-8 nanoparticles for improved breast cancer therapy: An in vitro and in vivo study. Colloids and Surfaces B: Biointerfaces, 2021, 203: 111759
https://doi.org/10.1016/j.colsurfb.2021.111759 pmid: 33892283
59 Y T Qin, H Peng, X W He, et al.. pH-responsive polymer-stabilized ZIF-8 nanocomposites for fluorescence and magnetic resonance dual-modal imaging-guided chemo-/photodynamic combinational cancer therapy. ACS Applied Materials & Interfaces, 2019, 11(37): 34268–34281
https://doi.org/10.1021/acsami.9b12641 pmid: 31454217
60 J Zhu, H Li, Z Xiong, et al.. Polyethyleneimine-coated manganese oxide nanoparticles for targeted tumor PET/MR imaging. ACS Applied Materials & Interfaces, 2018, 10(41): 34954–34964
https://doi.org/10.1021/acsami.8b12355 pmid: 30234287
61 Z Liu, N Shen, Z Tang, et al.. An eximious and affordable GSH stimulus-responsive poly(α-lipoic acid) nanocarrier bonding combretastatin A4 for tumor therapy. Biomaterials Science, 2019, 7(7): 2803–2811
https://doi.org/10.1039/C9BM00002J pmid: 31062006
62 C Lin, H He, Y Zhang, et al.. Acetaldehyde-modified-cystine functionalized Zr-MOFs for pH/GSH dual-responsive drug delivery and selective visualization of GSH in living cells. RSC Advances, 2020, 10(6): 3084–3091
https://doi.org/10.1039/C9RA05741B
63 J Li, Y Wang, S Sun, et al.. Disulfide bond-based self-crosslinked carbon-dots for turn-on fluorescence imaging of GSH in living cells. The Analyst, 2020, 145(8): 2982–2987
https://doi.org/10.1039/D0AN00071J pmid: 32124898
64 B Lei, M Wang, Z Jiang, et al.. Constructing redox-responsive metal-organic framework nanocarriers for anticancer drug delivery. ACS Applied Materials & Interfaces, 2018, 10(19): 16698–16706
https://doi.org/10.1021/acsami.7b19693 pmid: 29692177
65 Y Liu, C S Gong, Y Dai, et al.. In situ polymerization on nanoscale metal-organic frameworks for enhanced physiological stability and stimulus-responsive intracellular drug delivery. Biomaterials, 2019, 218: 119365–119375
https://doi.org/10.1016/j.biomaterials.2019.119365 pmid: 31344642
66 J Deng, K Wang, M Wang, et al.. Mitochondria targeted nanoscale zeolitic imidazole framework-90 for ATP imaging in live cells. Journal of the American Chemical Society, 2017, 139(16): 5877–5882
https://doi.org/10.1021/jacs.7b01229 pmid: 28385016
67 X R Song, S H Li, J Dai, et al.. Polyphenol-inspired facile construction of smart assemblies for ATP- and pH-responsive tumor MR/Optical imaging and photothermal therapy. Small, 2017, 13(20): 1603997
https://doi.org/10.1002/smll.201603997 pmid: 28383201
68 W H Chen, X Yu, A Cecconello, et al.. Stimuli-responsive nucleic acid-functionalized metal-organic framework nanoparticles using pH- and metal-ion-dependent DNAzymes as locks. Chemical Science, 2017, 8(8): 5769–5780
https://doi.org/10.1039/C7SC01765K pmid: 28989617
69 W H Chen, W C Liao, Y S Sohn, et al.. Stimuli-responsive nucleic acid-based polyacrylamide hydrogel-coated metal-organic framework nanoparticles for controlled drug release. Advanced Functional Materials, 2018, 28(8): 1705137
https://doi.org/10.1002/adfm.201705137
70 S S Wan, L Zhang, X Z Zhang. An ATP-regulated ion transport nanosystem for homeostatic perturbation therapy and sensitizing photodynamic therapy by autophagy inhibition of tumors. ACS Central Science, 2019, 5(2): 327–340
https://doi.org/10.1021/acscentsci.8b00822 pmid: 30834321
71 W Zuo, D Chen, Z Fan, et al.. Design of light/ROS cascade-responsive tumor-recognizing nanotheranostics for spatiotemporally controlled drug release in locoregional photo-chemotherapy. Acta Biomaterialia, 2020, 111: 327–340
https://doi.org/10.1016/j.actbio.2020.04.052 pmid: 32434075
72 W Lv, H Xia, L Zou, et al.. Yolk–shell structured Au nanorods@mesoporous silica for gas bubble driven drug release upon near-infrared light irradiation. Nanomedicine: Nanotechnology, Biology, and Medicine, 2021, 32: 102326
https://doi.org/10.1016/j.nano.2020.102326 pmid: 33166666
73 X Zhu, Q Su, W Feng, et al.. Anti-Stokes shift luminescent materials for bio-applications. Chemical Society Reviews, 2017, 46(4): 1025–1039
https://doi.org/10.1039/C6CS00415F pmid: 27966684
74 J Xu, A Gulzar, Y Liu, et al.. Integration of IR-808 sensitized upconversion nanostructure and MoS2 nanosheet for 808 nm NIR light triggered phototherapy and bioimaging. Small, 2017, 13(36): 17018141
https://doi.org/10.1002/smll.201701841 pmid: 28737290
75 M Lismont, L Dreesen, S Wuttke. Metal-organic framework nanoparticles in photodynamic therapy: Current status and perspectives. Advanced Functional Materials, 2017, 27(14): 1606314
https://doi.org/10.1002/adfm.201606314
76 J Park, Q Jiang, D Feng, et al.. Size-controlled synthesis of porphyrinic metal-organic framework and functionalization for targeted photodynamic therapy. Journal of the American Chemical Society, 2016, 138(10): 3518–3525
https://doi.org/10.1021/jacs.6b00007 pmid: 26894555
77 J Liu, Y Yang, W Zhu, et al.. Nanoscale metal-organic frameworks for combined photodynamic & radiation therapy in cancer treatment. Biomaterials, 2016, 97: 1–9
https://doi.org/10.1016/j.biomaterials.2016.04.034 pmid: 27155362
78 B Li, X Wang, L Chen, et al.. Ultrathin Cu-TCPP MOF nanosheets: A new theragnostic nanoplatform with magnetic resonance/near-infrared thermal imaging for synergistic phototherapy of cancers. Theranostics, 2018, 8(15): 4086–4096
https://doi.org/10.7150/thno.25433 pmid: 30128038
79 Y Wang, L Shi, D Ma, et al.. Tumor-activated and metal-organic framework assisted self-assembly of organic photosensitizers. ACS Nano, 2020, 14(10): 13056–13068
https://doi.org/10.1021/acsnano.0c04518 pmid: 33016697
80 D Yang, J Xu, G Yang, et al.. Metal-organic frameworks join hands to create an anti-cancer nanoplatform based on 808 nm light driving up-conversion nanoparticles. Chemical Engineering Journal, 2018, 344: 363–374
https://doi.org/10.1016/j.cej.2018.03.101
81 J Huang, Z Xu, Y Jiang, et al.. Metal organic framework-coated gold nanorod as an on-demand drug delivery platform for chemo-photothermal cancer therapy. Journal of Nanobiotechnology, 2021, 19(1): 219
https://doi.org/10.1186/s12951-021-00961-x pmid: 34281545
82 X Cai, Z Xie, B Ding, et al.. Monodispersed copper(I)-based nano metal-organic framework as a biodegradable drug carrier with enhanced photodynamic therapy efficacy. Advanced Science, 2019, 6(15): 1900848
https://doi.org/10.1002/advs.201900848 pmid: 31406677
83 X Cai, Y Jiang, M Lin, et al.. Ultrasound-responsive materials for drug/gene delivery. Frontiers in Pharmacology, 2020, 10: 1650
https://doi.org/10.3389/fphar.2019.01650 pmid: 32082157
84 X Cui, X Han, L Yu, et al.. Intrinsic chemistry and design principle of ultrasound-responsive nanomedicine. Nano Today, 2019, 28: 100773
https://doi.org/10.1016/j.nantod.2019.100773
85 M Ibrahim, R Sabouni, G A Husseini, et al.. Facile ultrasound-triggered release of calcein and doxorubicin from iron-based metal-organic frameworks. Journal of Biomedical Nanotechno-logy, 2020, 16(9): 1359–1369
https://doi.org/10.1166/jbn.2020.2972 pmid: 33419490
86 Y Zhou, M Wang, Z Dai. The molecular design of and challenges relating to sensitizers for cancer sonodynamic therapy. Materials Chemistry Frontiers, 2020, 4(8): 2223–2234
https://doi.org/10.1039/D0QM00232A
87 C Huang, S Ding, W Jiang, et al.. Glutathione-depleting nanoplatelets for enhanced sonodynamic cancer therapy. Nano-scale, 2021, 13(8): 4512–4518
https://doi.org/10.1039/D0NR08440A pmid: 33615325
88 X Pan, W Wang, Z Huang, et al.. MOF-derived double-layer hollow nanoparticles with oxygen generation ability for multimodal imaging-guided sonodynamic therapy. Angewandte Chemie International Edition, 2020, 59(32): 13557–13561
https://doi.org/10.1002/anie.202004894 pmid: 32374941
89 X Ke, X Song, N Qin, et al.. Rational synthesis of magnetic Fe3O4@MOF nanoparticles for sustained drug delivery. Journal of Porous Materials, 2019, 26(3): 813–818
https://doi.org/10.1007/s10934-018-0682-4
90 M Aghayi-Anaraki, V Safarifard. Fe3O4@MOF magnetic nanocomposites: Synthesis and applications. European Journal of Inorganic Chemistry, 2020, 2020(20): 1916–1937
https://doi.org/10.1002/ejic.202000012
91 Z Xiang, Y Qi, Y Lu, et al.. MOF-derived novel porous Fe3O4@C nanocomposites as smart nanomedical platforms for combined cancer therapy: Magnetic-triggered synergistic hyperthermia and chemotherapy. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2020, 8(37): 8671–8683
https://doi.org/10.1039/D0TB01021A pmid: 32856668
92 R Lin, W Yu, X Chen, et al.. Self-propelled micro/nanomotors for tumor targeting delivery and therapy. Advanced Healthcare Materials, 2021, 10(1): 2001212
https://doi.org/10.1002/adhm.202001212 pmid: 32975892
93 J Xu, S S Lee, H Seo, et al.. Quinic acid-conjugated nanoparticles enhance drug delivery to solid tumors via interactions with endothelial selectins. Small, 2018, 14(50): 1803601
https://doi.org/10.1002/smll.201803601 pmid: 30411856
94 J Park, Y Choi, H Chang, et al.. Alliance with EPR effect: Combined strategies to improve the EPR effect in the tumor microenvironment. Theranostics, 2019, 9(26): 8073–8090
https://doi.org/10.7150/thno.37198 pmid: 31754382
95 T Xue, C Xu, Y Wang, et al.. Doxorubicin-loaded nanoscale metal-organic framework for tumor-targeting combined chemotherapy and chemodynamic therapy. Biomaterials Science, 2019, 7(11): 4615–4623
https://doi.org/10.1039/C9BM01044K pmid: 31441464
96 P Liu, Y Zhou, X Shi, et al.. A cyclic nano-reactor achieving enhanced photodynamic tumor therapy by reversing multiple resistances. Journal of Nanobiotechnology, 2021, 19(1): 149
https://doi.org/10.1186/s12951-021-00893-6 pmid: 34020663
97 J Yan, C Liu, Q Wu, et al.. Mineralization of pH-sensitive doxorubicin prodrug in ZIF-8 to enable targeted delivery to solid tumors. Analytical Chemistry, 2020, 92(16): 11453–11461
https://doi.org/10.1021/acs.analchem.0c02599 pmid: 32664723
98 W Xu, Y Lou, W Chen, et al.. Folic acid decorated metal-organic frameworks loaded with doxorubicin for tumor-targeted chemotherapy of osteosarcoma. Biomedical Engineerin/Biomedizinische Technik, 2020, 65(2): 229–236
https://doi.org/10.1515/bmt-2019-0056
99 A Samui, K Pal, P Karmakar, et al.. In situ synthesized lactobionic acid conjugated NMOFs, a smart material for imaging and targeted drug delivery in hepatocellular carcinoma. Materials Science and Engineering C, 2019, 98: 772–781
https://doi.org/10.1016/j.msec.2019.01.032 pmid: 30813083
100 H Zhang, Q Zhang, C Liu, et al.. Preparation of a one-dimensional nanorod/metal organic framework Janus nanoplatform via side-specific growth for synergistic cancer therapy. Biomaterials Science, 2019, 7(4): 1696–1704
https://doi.org/10.1039/C8BM01591K pmid: 30747179
101 Y He, T Xiong, S He, et al.. Pulmonary targeting crosslinked cyclodextrin metal-organic frameworks for lung cancer therapy. Advanced Functional Materials, 2021, 31(3): 2004550
https://doi.org/10.1002/adfm.202004550
102 Y Zhang, L Lin, L Liu, et al.. Positive feedback nanoamplifier responded to tumor microenvironments for self-enhanced tumor imaging and therapy. Biomaterials, 2019, 216: 119255–119263
https://doi.org/10.1016/j.biomaterials.2019.119255 pmid: 31229855
103 K Kim, S Lee, E Jin, et al.. MOF × biopolymer: Collaborative combination of metal-organic framework and biopolymer for advanced anticancer therapy. ACS Applied Materials & Interfaces, 2019, 11(31): 27512–27520
https://doi.org/10.1021/acsami.9b05736 pmid: 31293157
104 M Wu, X Liu, H Bai, et al.. Surface-layer protein-enhanced immunotherapy based on cell membrane-coated nanoparticles for the effective inhibition of tumor growth and metastasis. ACS Applied Materials & Interfaces, 2019, 11(10): 9850–9859
https://doi.org/10.1021/acsami.9b00294 pmid: 30788951
105 Z Chai, X Hu, W Lu. Cell membrane-coated nanoparticles for tumor-targeted drug delivery. Science China Materials, 2017, 60(6): 504–510
https://doi.org/10.1007/s40843-016-5163-4
106 X Wan, L Song, W Pan, et al.. Tumor-targeted cascade nanoreactor based on metal-organic frameworks for synergistic ferroptosis-starvation anticancer therapy. ACS Nano, 2020, 14(9): 11017–11028
https://doi.org/10.1021/acsnano.9b07789 pmid: 32786253
107 E A Owens, M Henary, G El Fakhri, et al.. Tissue-specific near-infrared fluorescence imaging. Accounts of Chemical Research, 2016, 49(9): 1731–1740
https://doi.org/10.1021/acs.accounts.6b00239 pmid: 27564418
108 J Liu, Z Liu, D Wu. Multifunctional hypoxia imaging nanoparticles: multifunctional tumor imaging and related guided tumor therapy. International Journal of Nanomedicine, 2019, 14: 707–719
https://doi.org/10.2147/IJN.S192048 pmid: 30705587
109 J Sun, J Wang, W Hu, et al.. Camouflaged gold nanodendrites enable synergistic photodynamic therapy and NIR biowindow II photothermal therapy and multimodal imaging. ACS Applied Materials & Interfaces, 2021, 13(9): 10778–10795
https://doi.org/10.1021/acsami.1c01238 pmid: 33646767
110 Z Ouyang, D Li, Z Xiong, et al.. Antifouling dendrimer-entrapped copper sulfide nanoparticles enable photoacoustic imaging-guided targeted combination therapy of tumors and tumor metastasis. ACS Applied Materials & Interfaces, 2021, 13(5): 6069–6080
https://doi.org/10.1021/acsami.0c21620 pmid: 33501834
111 G Zhou, Y S Wang, Z Jin, et al.. Porphyrin-palladium hydride MOF nanoparticles for tumor-targeting photoacoustic imaging-guided hydrogenothermal cancer therapy. Nanoscale Horizons, 2019, 4(5): 1185–1193
https://doi.org/10.1039/C9NH00021F
112 Y Wang, W Wu, D Mao, et al.. Metal-organic framework assisted and tumor microenvironment modulated synergistic image-guided photo-chemo therapy. Advanced Functional Materials, 2020, 30(28): 2002431
https://doi.org/10.1002/adfm.202002431
113 Y Chen, Z H Li, P Pan, et al.. Tumor-microenvironment-triggered ion exchange of a metal-organic framework hybrid for multimodal imaging and synergistic therapy of tumors. Advanced Materials, 2020, 32(24): 2001452
https://doi.org/10.1002/adma.202001452 pmid: 32374492
114 M Peller, K Böll, A Zimpel, et al.. Metal-organic framework nanoparticles for magnetic resonance imaging. Inorganic Che-mistry Frontiers, 2018, 5(8): 1760–1779
https://doi.org/10.1039/C8QI00149A
115 S M McLeod, L Robison, G Parigi, et al.. Maximizing magnetic resonance contrast in Gd(III) nanoconjugates: Investigation of proton relaxation in zirconium metal-organic frameworks. ACS Applied Materials & Interfaces, 2020, 12(37): 41157–41166
https://doi.org/10.1021/acsami.0c13571 pmid: 32852198
116 J Yao, Y Liu, J Wang, et al.. On-demand CO release for amplification of chemotherapy by MOF functionalized magnetic carbon nanoparticles with NIR irradiation. Biomaterials, 2019, 195: 51–62
https://doi.org/10.1016/j.biomaterials.2018.12.029 pmid: 30610993
117 A Ebrahimpour, N Riahi Alam, P Abdolmaleki, et al.. Magnetic metal-organic framework based on zinc and 5-aminolevulinic acid: MR imaging and brain tumor therapy. Journal of Inorganic and Organometallic Polymers and Materials, 2021, 31(3): 1208–1216
https://doi.org/10.1007/s10904-020-01782-5
118 H Zhou, M Qi, J Shao, et al.. Manganese oxide/metal-organic frameworks-based nanocomposites for tumr micro-environment sensitive 1H/19F dual-mode magnetic resonance imaging in vivo. Journal of Organometallic Chemistry, 2021, 933: 121652
https://doi.org/10.1016/j.jorganchem.2020.121652
119 C Guo, S Xu, A Arshad, et al.. A pH-responsive nanoprobe for turn-on 19F-magnetic resonance imaging. Chemical Communications, 2018, 54(70): 9853–9856
https://doi.org/10.1039/C8CC06129G pmid: 30112535
120 W Zhu, M Chen, Y Liu, et al.. A dual factor activated metal-organic framework hybrid nanoplatform for photoacoustic imaging and synergetic photo-chemotherapy. Nanoscale, 2019, 11(43): 20630–20637
https://doi.org/10.1039/C9NR06349H pmid: 31641722
121 Y Pu, Y Zhu, Z Qiao, et al.. A Gd-doped polydopamine (PDA)-based theranostic nanoplatform as a strong MR/PA dual-modal imaging agent for PTT/PDT synergistic therapy. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2021, 9(7): 1846–1857
https://doi.org/10.1039/D0TB02725A pmid: 33527969
122 S S Wan, Q Cheng, X Zeng, et al.. A Mn(III)-sealed metal-organic framework nanosystem for redox-unlocked tumor theranostics. ACS Nano, 2019, 13(6): 6561–6571
https://doi.org/10.1021/acsnano.9b00300 pmid: 31136707
123 Y Zhu, N Xin, Z Qiao, et al.. Bioactive MOFs based theranostic agent for highly effective combination of multimodal imaging and chemo-phototherapy. Advanced Healthcare Materials, 2020, 9(14): 2000205
https://doi.org/10.1002/adhm.202000205 pmid: 32548979
124 W Cai, H Gao, C Chu, et al.. Engineering phototheranostic nanoscale metal-organic frameworks for multimodal imaging-guided cancer therapy. ACS Applied Materials & Interfaces, 2017, 9(3): 2040–2051
https://doi.org/10.1021/acsami.6b11579 pmid: 28032505
125 X Sun, G He, C Xiong, et al.. One-pot fabrication of hollow porphyrinic MOF nanoparticles with ultrahigh drug loading toward controlled delivery and synergistic cancer therapy. ACS Applied Materials & Interfaces, 2021, 13(3): 3679–3693
https://doi.org/10.1021/acsami.0c20617 pmid: 33464038
[1] Xin LIU, Xiangling REN, Longfei TAN, Wenna GUO, Zhongbing HUANG, Xianwei MENG. Preparation and enhanced properties of ZrMOF@CdTe nanoparticles with high-density quantum dots[J]. Front. Mater. Sci., 2020, 14(2): 155-162.
[2] Maria COROŞ, Florina POGĂCEAN, Lidia MĂGERUŞAN, Crina SOCACI, Stela PRUNEANU. A brief overview on synthesis and applications of graphene and graphene-based nanomaterials[J]. Front. Mater. Sci., 2019, 13(1): 23-32.
[3] Honghong ZOU, Lei WANG, Chenghui ZENG, Xiaolei GAO, Qingqing WANG, Shengliang ZHONG. Rare-earth coordination polymer micro/nanomaterials: Preparation, properties and applications[J]. Front. Mater. Sci., 2018, 12(4): 327-347.
[4] Ramanathan SARASWATHY. Electrochemical capacitance of nanostructured ruthenium-doped tin oxide Sn1--xRuxO2 by the microemulsion method[J]. Front. Mater. Sci., 2017, 11(4): 385-394.
[5] V. CHELLASAMY, P. THANGADURAI. Structural and electrochemical investigations of nanostructured NiTiO3 in acidic environment[J]. Front. Mater. Sci., 2017, 11(2): 162-170.
[6] Diah SUSANTI, Rizky Narendra Dwi WIBAWA, Lucky TANANTA, Hariyati PURWANINGSIH, Rindang FAJARIN, George Endri KUSUMA. The effect of calcination temperature on the capacitive properties of WO3-based electrochemical capacitors synthesized via a sol--gel method[J]. Front Mater Sci, 2013, 7(4): 370-378.
[7] Andris SUTKA, Gundars MEZINSKIS. Sol–gel auto-combustion synthesis of spinel-type ferrite nanomaterials[J]. Front Mater Sci, 2012, 6(2): 128-141.
[8] Lan-Zheng REN, Jin-Xiu WANG. A simple hydrothermal route to fabrication of single-crystalline silver nanoplates using poly(vinyl pyrrolidone)[J]. Front Mater Sci Chin, 2010, 4(4): 407-410.
[9] Lan-Zheng REN, Jin-Xiu WANG. Facile one-pot preparation of silver nanowires using an alcohol ionic liquid[J]. Front Mater Sci Chin, 2010, 4(4): 398-401.
[10] En-Rong LI, Qian-Jun ZHANG, Wei WANG, Qing-Wen ZHU, Long BA, . Fabrication of mesoporous silica/carbon black nanospheres and load-sensitive conducting rubber nanocomposites[J]. Front. Mater. Sci., 2010, 4(1): 52-56.
[11] F. WATARI, T. AKASAKA, Xiaoming LI, M. UO, A. YOKOYAMA. Proliferation of osteoblast cells on nanotubes[J]. Front Mater Sci Chin, 2009, 3(2): 169-173.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed