4.8 Article

Self-assembled green tea polyphenol-based coordination nanomaterials to improve chemotherapy efficacy by inhibition of carbonyl reductase 1

Journal

BIOMATERIALS
Volume 210, Issue -, Pages 62-69

Publisher

ELSEVIER SCI LTD
DOI: 10.1016/j.biomaterials.2019.04.032

Keywords

Chemotherapy; Epigallocatechin-3-O-Gallate (EGCG); Positron emission tomography; Doxorubicin; Carbonyl reductase 1 (CBR1); Polyphenols

Funding

  1. National Science Foundation Committee of Anhui Province [1808085MH256]
  2. Innovation Team of Pharmaceutical Biotechnology [2016KYTD01]
  3. Key Project of Anhui Educational Committee [KJ2013A262, KJ2015A220, gxfxZD2016266]
  4. Quality Project of AnHui Education Department-The Plan of ChuangKe Laboratory [2015ckjh109]
  5. Research Platform Project of Suzhou University [2016kyf08]
  6. intramural research program of Faculty of Health Sciences, University of Macau
  7. Science and Technology Development Fund (FDCT) of Macao SAR [FDCT 0109/2018/A3]
  8. Start-up Research Grant (SRG) of University of Macau [SRG2018-00130-FHS]
  9. Intramural Research Program (IRP) of the National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH)
  10. China Scholarship Council (CSC)
  11. NATIONAL INSTITUTE OF BIOMEDICAL IMAGING AND BIOENGINEERING [ZIAEB000073] Funding Source: NIH RePORTER

Ask authors/readers for more resources

Nanomedicine has become a promising approach to improve cancer chemotherapy. It remains a major challenge how to enhance anti-drug efficacy and reduce side effects of anti-cancer drugs. Herein, we report a self-assembled nanoplatform (FDEP NPs) by integration of doxorubicin (DOX) and epigallocatechin-3-O-gallate (EGCG) with the help of coordination between Fe3+ ions and polyphenols. The EGCG from FDEP NPs could inhibit the expression of carbonyl reductase 1 (CBR1) protein and thereby inhibit the doxorubicinol (DOXOL) generation from DOX both in vitro and in vivo, thus the efficacy of DOX to cancerous cells is improved significantly. More importantly, the FDEP NPs could reduce cardiac toxicity and the DOX mediated toxicity to blood cells due to the repression of DOXOL production. Moreover, the blood half-life of FDEP NPs is longer than 23 has determined by positron emission tomography (PET) imaging of biodistribution of radiolabelled NPs and HPLC measurement of plasma level of DOX, ensuring high tumor accumulation of FDEP NPs by enhanced permeability and retention (EPR) effect. The FDEP NPs also exhibited much improved antitumor effect over free drugs. Our work sheds new light on the engineering of nanomaterials for combination chemotherapy and may find unique clinical applications in the near future.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available