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 (4) : 553-566    https://doi.org/10.1007/s11706-021-0576-2
RESEARCH ARTICLE
A nanosystem of copper(II)-disulfiram for cancer treatment with high efficacy and few side effects
Liping ZHAO1, Xiaoxia WANG1, Mingxia JIANG1, Xinghan WU2, Mogen ZHANG3, Xiuwen GUAN1,4,5, Jinlong MA1,4,5(), Weifen ZHANG1,4,5()
1. College of Pharmacy, Weifang Medical University, Weifang 261053, China
2. Department of Pathology, Weifang Medical University, Weifang 261053, China
3. School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
4. Collaborative Innovation Center for Target Drug Delivery System, Weifang Medical University, Weifang 261053, China
5. Shandong Engineering Research Center for Smart Materials and Regenerative Medicine, Weifang Medical University, Weifang 261053, China
 Download: PDF(6904 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Developing chemotherapy drugs with high efficacy and few side effects has been a bottleneck problem that requires an efficient solution. The active cancer treatment ingredient disulfiram (DSF), inspired by the copper(II) diethyldithiocarbamate complex (CuET), can be used in a one-pot synthesis method to construct a CuET delivery nanosystem (CuET-ZIFCu@HA). Due to the high biocompatibility, targeting of CD44 overexpressed cancer cells, and acid response of zeolitic imidazolate framework (ZIF) materials of hyaluronic acid (HA), we realized that CuET-ZIFCu@HA could become an effective and highly selective cancer treatment. Both in vivo and in vitro experiments have demonstrated that CuET-ZIFCu@HA has robust anti-tumor properties without evident side effects. This research provided a promising strategy for DSF nanosystems that involves simple preparation and high efficacy, both of which are key to reusing DSF in cancer treatment.

Keywords disulfiram      copper(II) diethyldithiocarbamate complex      zeolitic imidazolate framework      targeted therapy     
Corresponding Author(s): Jinlong MA,Weifen ZHANG   
Online First Date: 10 November 2021    Issue Date: 28 December 2021
 Cite this article:   
Liping ZHAO,Xiaoxia WANG,Mingxia JIANG, et al. A nanosystem of copper(II)-disulfiram for cancer treatment with high efficacy and few side effects[J]. Front. Mater. Sci., 2021, 15(4): 553-566.
 URL:  
https://academic.hep.com.cn/foms/EN/10.1007/s11706-021-0576-2
https://academic.hep.com.cn/foms/EN/Y2021/V15/I4/553
Fig.1  Scheme 1 Schematic illustration of CuET delivery nanosystem (CuET-ZIFCu@HA) by one-pot synthesis method and in vivo efficacy evaluation for cancer.
Fig.2  (a) DLS profile of CuET-ZIFCu and CuET-ZIFCu@HA. (b) Zeta potentials of ZIFCu, CuET-ZIFCu, and CuET-ZIFCu@HA; results are presented as mean ± SD, n = 3. (c) The UV–Vis absorption spectra of ZIFCu, CuET-ZIFCu, and CuET-ZIFCu@HA. (d) TEM images of CuET-ZIFCu and CuET-ZIFCu@HA. (e) Mapping images of CuET-ZIFCu@HA.
Fig.3  The kinetics of drug release from (a) CuET-ZIFCu and (b) CuET-ZIFCu@HA under various buffer conditions; results are presented as mean ± SD, n = 3. Hemolysis activity of (c) CuET-ZIFCu and (d) CuET-ZIFCu@HA; results are presented as mean ± SD, n = 3. (e) The LIVE/DEAD assay of L929 cells after different treatments at the CuET concentration of 1.5 μg·mL−1. (f) Apoptosis rates of L929 cells by flow cytometry after 24 h co-incubation with CuET, CuET-ZIFCu, and CuET-ZIFCu@HA (concentration of 1.5 μg·mL−1).
Fig.4  (a) Cell viability of 231 cells after treatment with different concentrations of CuET; results are presented as mean ± SD. (b) The LIVE/DEAD assay of 231 cells after different treatments at the CuET concentration of 1.5 μg·mL−1. (c) Apoptosis rates of 231 cells by flow cytometry after 24 h co-incubation with CuET (concentration of 1.5 μg·mL−1).
Fig.5  ROS images and quantitative analyses of 231 cells after treatment with CuET at the concentrations of (a) 1 μg·mL−1 and (b) 1.5 μg·mL−1.
Fig.6  (a) Biodistribution of Cy5.5 and CuET-ZIFCu@HA-Cy5.5 in tumor-bearing nude mice at different times. (b) Biodistribution of Cy5.5 and CuET-ZIFCu@HA-Cy5.5 in the major organs (tumor, heart, liver, spleen, lung, and kidney) were harvested at 6 h. (c) Semi-quantitative analysis of the biodistribution profiles of Cy5.5 and CuET-ZIFCu@HA-Cy5.5 in tumor-bearing mice.
Fig.7  (a) Tumor growth curves after treatment with PBS, ZIFCu, CuET-ZIFCu, and CuET-ZIFCu@HA. (b) Weights of mice treated with PBS, ZIFCu, CuET-ZIFCu, and CuET-ZIFCu@HA during the 14-d treatment. (c) Weights of tumors in the groups of PBS, ZIFCu, CuET-ZIFCu, and CuET-ZIFCu@HA. (d) Images of tumors after treatment with PBS, ZIFCu, CuET-ZIFCu, and CuET-ZIFCu@HA. (e) H&E section of tumor after treatment at day 14. (f) H&E section of the heart, liver, spleen, lung, and kidney after treatment for 14 d. The data expressed as the arithmetic mean ± SD. ***p<0.001.
  Fig. S1 The size of ZIFCu.
  Fig. S2 Stability of CuET-ZIFCu and CuET-ZIFCu@HA in RPMI over a period of one week.
c(CuET)/(mg·mL−1) EE/% DL/%
CuET-ZIFCu CuET-ZIFCu@HA
1 75.68±0.80 15.33±0.14 10.57±0.08
1.2 76.00±0.40 19.90±0.08 12.47±0.05
1.4 75.95±0.15 20.28±0.03 14.25±0.05
1.6 74.00±0.28 22.07±0.05 15.61±0.03
1.8 72.96±0.12 23.90±0.03 17.03±0.02
2.0 70.66±0.11 25.27±0.03 18.09±0.02
  EE and DL values in CuET-ZIFCu and CuET-ZIFCu@HA
1 L Qiu, T Chen, I Öçsoy, et al.. A cell-targeted, size-photocontrollable, nuclear-uptake nanodrug delivery system for drug-resistant cancer therapy. Nano Letters, 2015, 15(1): 457–463
https://doi.org/10.1021/nl503777s pmid: 25479133
2 J Ma, Z Hu, W Wang, et al.. pH-sensitive reversible programmed targeting strategy by the self-assembly/disassembly of gold nanoparticles. ACS Applied Materials & Interfaces, 2017, 9(20): 16767–16777
https://doi.org/10.1021/acsami.7b00687 pmid: 28489342
3 L Zhang, S S Wan, C X Li, et al.. An adenosine triphosphate-responsive autocatalytic fenton nanoparticle for tumor ablation with self-supplied H2O2 and acceleration of Fe(III)/Fe(II) conversion. Nano Letters, 2018, 18(12): 7609–7618
https://doi.org/10.1021/acs.nanolett.8b03178 pmid: 30383966
4 J Wang, X Wu, P Shen, et al.. Applications of inorganic nanomaterials in photothermal therapy based on combinational cancer treatment. International Journal of Nanomedicine, 2020, 15: 1903–1914
https://doi.org/10.2147/IJN.S239751 pmid: 32256067
5 Y Ding, H Xu, C Xu, et al.. A nanomedicine fabricated from gold nanoparticles-decorated metal-organic framework for cascade chemo/chemodynamic cancer therapy. Advanced Science, 2020, 7(17): 2001060
https://doi.org/10.1002/advs.202001060 pmid: 32995124
6 Z Gu, S Zhu, L Yan, et al.. Graphene-based smart platforms for combined cancer therapy. Advanced Materials, 2019, 31(9): 1800662
https://doi.org/10.1002/adma.201800662 pmid: 30039878
7 X Bao, Y Yuan, J Chen, et al.. In vivo theranostics with near-infrared-emitting carbon dots-highly efficient photothermal therapy based on passive targeting after intravenous administration. Light: Science & Applications, 2018, 7(1): 91
https://doi.org/10.1038/s41377-018-0090-1 pmid: 30479757
8 D Majera, Z Skrott, J Bouchal, et al.. Targeting genotoxic and proteotoxic stress-response pathways in human prostate cancer by clinically available PARP inhibitors, vorinostat and disulfiram. Prostate, 2019, 79(4): 352–362
https://doi.org/10.1002/pros.23741 pmid: 30499118
9 Z Skrott, M Mistrik, K K Andersen, et al.. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature, 2017, 552(7684): 194–199
https://doi.org/10.1038/nature25016 pmid: 29211715
10 L Zhou, L Yang, C Yang, et al.. Membrane loaded copper oleate PEGylated liposome combined with disulfiram for improving synergistic antitumor effect in vivo. Pharmaceutical Research, 2018, 35(7): 147
https://doi.org/10.1007/s11095-018-2414-5 pmid: 29796711
11 Z Skrott, D Majera, J Gursky, et al.. Disulfiram’s anti-cancer activity reflects targeting NPL4, not inhibition of aldehyde dehydrogenase. Oncogene, 2019, 38(40): 6711–6722
https://doi.org/10.1038/s41388-019-0915-2 pmid: 31391554
12 K Wang, T Michelakos, B Wang, et al.. Targeting cancer stem cells by disulfiram and copper sensitizes radioresistant chondrosarcoma to radiation. Cancer Letters, 2021, 505: 37–48
https://doi.org/10.1016/j.canlet.2021.02.002 pmid: 33582212
13 L Zhang, B Tian, Y Li, et al.. A copper-mediated disulfiram-loaded pH-triggered PEG-shedding TAT peptide-modified lipid nanocapsules for use in tumor therapy. ACS Applied Materials & Interfaces, 2015, 7(45): 25147–25161
https://doi.org/10.1021/acsami.5b06488 pmid: 26501354
14 X Peng, Q Pan, B Zhang, et al.. Highly stable, coordinated polymeric nanoparticles loading copper(II) diethyldithiocarbamate for combinational chemo/chemodynamic therapy of cancer. Biomacromolecules, 2019, 20(6): 2372–2383
https://doi.org/10.1021/acs.biomac.9b00367 pmid: 31117352
15 Q Yang, Y Yao, K Li, et al.. An updated review of disulfiram: Molecular targets and strategies for cancer treatment. Current Pharmaceutical Design, 2019, 25(30): 3248–3256
https://doi.org/10.2174/1381612825666190816233755 pmid: 31419930
16 Y Yang, F Wang, Q Yang, et al.. Hollow metal-organic framework nanospheres via emulsion-based interfacial synthesis and their application in size-selective catalysis. ACS Applied Materials & Interfaces, 2014, 6(20): 18163–18171
https://doi.org/10.1021/am505145d pmid: 25247890
17 D Wang, J Zhou, R Chen, et al.. Controllable synthesis of dual-MOFs nanostructures for pH-responsive artemisinin delivery, magnetic resonance and optical dual-model imaging-guided chemo/photothermal combinational cancer therapy. Biomaterials, 2016, 100: 27–40
https://doi.org/10.1016/j.biomaterials.2016.05.027 pmid: 27240160
18 D Liu, J Wan, G Pang, et al.. Hollow metal-organic-framework micro/nanostructures and their derivatives: Emerging multifunctional materials. Advanced Materials, 2019, 31(38): 1803291
https://doi.org/10.1002/adma.201803291 pmid: 30548351
19 S Zhang, X Pei, H Gao, et al.. Metal-organic framework-based nanomaterials for biomedical applications. Chinese Chemical Letters, 2020, 31(5): 1060–1070
https://doi.org/10.1016/j.cclet.2019.11.036
20 Q Wu, M Li, L Tan, et al.. A tumor treatment strategy based on biodegradable BSA@ZIF-8 for simultaneously ablating tumors and inhibiting infection. Nanoscale Horizons, 2018, 3(6): 606–615
https://doi.org/10.1039/C8NH00113H pmid: 32254113
21 F Shu, D Lv, X L Song, et al.. Fabrication of a hyaluronic acid conjugated metal organic framework for targeted drug delivery and magnetic resonance imaging. RSC Advances, 2018, 8(12): 6581–6589
https://doi.org/10.1039/c7ra12969f
22 S Dissegna, K Epp, W R Heinz, et al.. Defective metal-organic frameworks. Advanced Materials, 2018, 30(37): 1704501
https://doi.org/10.1002/adma.201704501 pmid: 29363822
23 H Zheng, Y Zhang, L Liu, et al.. One-pot synthesis of metal-organic frameworks with encapsulated target molecules and their applications for controlled drug delivery. Journal of the American Chemical Society, 2016, 138(3): 962–968
https://doi.org/10.1021/jacs.5b11720 pmid: 26710234
24 W Zhang, J Lu, X Gao, et al.. Enhanced photodynamic therapy by reduced levels of intracellular glutathione obtained by employing a nano-MOF with CuII as the active center. Angewandte Chemie International Edition, 2018, 57(18): 4891–4896
https://doi.org/10.1002/anie.201710800 pmid: 29451722
25 N C Burtch, J Heinen, T D Bennett, et al.. Mechanical properties in metal-organic frameworks: Emerging opportunities and challenges for device functionality and technological applications. Advanced Materials, 2018, 30(37): 1704124
https://doi.org/10.1002/adma.201704124 pmid: 29149545
26 Z Luo, Y Dai, H Gao. Development and application of hyaluronic acid in tumor targeting drug delivery. Acta Pharmaceutica Sinica B, 2019, 9(6): 1099–1112
https://doi.org/10.1016/j.apsb.2019.06.004 pmid: 31867159
27 R Liu, W Xiao, C Hu, et al.. Theranostic size-reducible and no donor conjugated gold nanocluster fabricated hyaluronic acid nanoparticle with optimal size for combinational treatment of breast cancer and lung metastasis. Journal of Controlled Release, 2018, 278: 127–139
https://doi.org/10.1016/j.jconrel.2018.04.005 pmid: 29630985
28 Z Wang, Z Chen, Z Liu, et al.. A multi-stimuli responsive gold nanocage-hyaluronic platform for targeted photothermal and chemotherapy. Biomaterials, 2014, 35(36): 9678–9688
https://doi.org/10.1016/j.biomaterials.2014.08.013 pmid: 25176062
29 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
30 L Su, Q Wu, L Tan, et al.. High biocompatible ZIF-8 coated by ZrO2 for chemo-microwave thermal tumor synergistic therapy. ACS Applied Materials & Interfaces, 2019, 11(11): 10520–10531
https://doi.org/10.1021/acsami.8b22177 pmid: 30801175
31 Z Tang, Y Liu, M He, et al.. Chemodynamic therapy: Tumour microenvironment-mediated fenton and fenton-like reactions. Angewandte Chemie International Edition, 2019, 58(4): 946–956
https://doi.org/10.1002/anie.201805664 pmid: 30048028
32 W Wu, L Yu, Q Jiang, et al.. Enhanced tumor-specific disulfiram chemotherapy by in situ Cu2+ chelation-initiated nontoxicity-to-toxicity transition. Journal of the American Chemical Society, 2019, 141(29): 11531–11539
https://doi.org/10.1021/jacs.9b03503 pmid: 31251050
33 Y Peng, P Liu, Y Meng, et al.. Nanoscale copper(II)–diethyldithiocarbamate coordination polymer as a drug self-delivery system for highly robust and specific cancer therapy. Molecular Pharmaceutics, 2020, 17(8): 2864–2873
https://doi.org/10.1021/acs.molpharmaceut.0c00284 pmid: 32551674
34 Y Hua, X Lv, Y Cai, et al.. Highly selective and reproducible electroanalysis for histidine in blood with turn-on responses at a potential approaching zero using tetrahedral copper metal organic frameworks. Chemical Communications, 2019, 55(9): 1271–1274
https://doi.org/10.1039/C8CC09562K pmid: 30632572
35 Y Liu, P Bhattarai, Z Dai, et al.. Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chemical Society Reviews, 2019, 48(7): 2053–2108
https://doi.org/10.1039/C8CS00618K pmid: 30259015
36 M Zhang, X Guo, M Wang, et al.. Tumor microenvironment-induced structure changing drug/gene delivery system for overcoming delivery-associated challenges. Journal of Controlled Release, 2020, 323: 203–224
https://doi.org/10.1016/j.jconrel.2020.04.026 pmid: 32320817
37 Y Wang, W Wu, J Liu, et al.. Cancer-cell-activated photodynamic therapy assisted by Cu(II)-based metal-organic framework. ACS Nano, 2019, 13(6): 6879–6890
https://doi.org/10.1021/acsnano.9b01665 pmid: 31194910
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed