Fabricating polydopamine-coated MoSe 2 -wrapped hollow mesoporous silica nanoplatform for controlled drug release and chemo-photothermal therapy

Integration of several types of therapeutic agents into one nanoplatform to enhance treatment efficacy is being more widely used for cancer therapy. Herein, a biocompatible polydopamine (PDA)-coated MoSe -wrapped doxorubicin (DOX)-loaded hollow mesoporous silica nanoparticles (HMSNs) nanoplatform (P...

Full description

Saved in:
Bibliographic Details
Published inInternational journal of nanomedicine Vol. 13; pp. 7607 - 7621
Main Authors Chai, Song, Kan, Shifeng, Sun, Ran, Zhou, Ruijuan, Sun, Yi, Chen, Wenhua, Yu, Bo
Format Journal Article
LanguageEnglish
Published New Zealand 01.11.2018
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Integration of several types of therapeutic agents into one nanoplatform to enhance treatment efficacy is being more widely used for cancer therapy. Herein, a biocompatible polydopamine (PDA)-coated MoSe -wrapped doxorubicin (DOX)-loaded hollow mesoporous silica nanoparticles (HMSNs) nanoplatform (PM@HMSNs-DOX) was fabricated for dual-sensitive drug release and chemo-photothermal therapy for enhancing the therapeutic effects on breast cancer. The HMSNs were obtained by a "structural difference-based selective etching" strategy and served as the drug carrier, exhibiting a high DOX loading capacity of 427 mg/g HMSNs-NH , and then wrapped with PDA-coated MoSe layer to form PM@HMSNs-DOX. Various techniques proved the successful fabrication of the nanocomposites. The formed PM@HMSNs-DOX nanocomposites exhibited good biocompatibility, good stability, and super-additive photothermal conversion efficiency due to the cooperation of MoSe and PDA. Simultaneously, the pH/near-infrared-responsive drug release profile was observed, which could enhance the synergistic therapeutic anticancer effect. The antitumor effects of PM@HMSNs-DOX were evaluated both in vitro and in vivo, demonstrating that the synergistic therapeutic efficacy was significantly superior to any monotherapy. Also, in vivo pharmacokinetics studies showed that PM@HMSNs-DOX had a much longer circulation time than free DOX. In addition, in vitro and in vivo toxicity studies certified that PM@HMSNs are suitable as biocompatible agents. Our nanoplatform loaded with DOX displays pH/near-infrared-induced chemotherapy and excellent photothermal therapy, which hold great potential for cancer treatment.
ISSN:1178-2013
1178-2013
DOI:10.2147/IJN.S181681