Reduction/photo dual-responsive polymeric prodrug nanoparticles for programmed siRNA and doxorubicin delivery

2018 ◽  
Vol 6 (6) ◽  
pp. 1457-1468 ◽  
Author(s):  
Ming Wu ◽  
Jiong Li ◽  
Xinyi Lin ◽  
Zuwu Wei ◽  
Da Zhang ◽  
...  

A reduction/photo dual-responsive nanosystem for programmed P-gp siRNA and doxorubicin release was designed to optimize the efficacy of chemotherapy against multidrug resistance of cancer cells.

2015 ◽  
Vol 17 (1) ◽  
pp. 384-398 ◽  
Author(s):  
Smita Kashyap ◽  
Nitesh Singh ◽  
Bapurao Surnar ◽  
Manickam Jayakannan

2021 ◽  
Author(s):  
Yupei Ma ◽  
Du Li ◽  
Yunchao Xiao ◽  
Zhijun OuYang ◽  
Mingwu Shen ◽  
...  

Conventional cancer chemotherapy is facing difficulties in improving the bioavailability, overcoming the severe adverse side effect of chemotherapeutics and reversing the multidrug resistance of cancer cells. To address these challenges,...


2003 ◽  
Vol 278 (28) ◽  
pp. 25285-25288 ◽  
Author(s):  
Suniti Misra ◽  
Shibnath Ghatak ◽  
Alexandra Zoltan-Jones ◽  
Bryan P. Toole

2021 ◽  
Vol 11 (5) ◽  
pp. 618-626
Author(s):  
Yali Liu ◽  
Changpeng Hu ◽  
Min Zhou ◽  
Qian Zhang ◽  
Qin Tang ◽  
...  

Doxorubicin (DOX) is widely used as a traditional chemotherapy drug in tumor treatment, but its dose-dependent side effects make it susceptible to acquired resistance. CXCR4 is a chemokine receptor that has high expression in many cancers, including lung cancer. In this work, we studied the possibility of using CXCR4 antagonist, AMD3100, as a targeting molecule to targeted delivery of DOX to CXCR4 expressing lung cancer cells through conjugated gold nanoparticles (Au NPs). DOX was intercalated inside the pH-responsive doublestrand DNA (dsDNA) and then conveniently loaded onto the Au NPs. The CXCR4 antagonist, AMD3100, was bonded with LA-PEG, and then conjugated to the surface of Au-S bond. The doxorubicin release from AuNPs@DOX@AMD3100 NPs was in a pH-dependent model, and specificity of AuNPs@DOX@AMD3100 nanoparticle was verified by using free DOX and Au@DOX NPs as control. Results in this work not only confirmed the possibility of using AMD3100 as a targeting ligand for tumor-targeted treatment, but also suggested that the non-toxic Au NPs is a prospect nanocarrier for target design of cancer therapy.


2020 ◽  
Vol 19 ◽  
pp. 153303382094580
Author(s):  
Ting Zhan ◽  
Xiaoli Chen ◽  
Xia Tian ◽  
Zheng Han ◽  
Meng Liu ◽  
...  

Background: Pancreatic cancer is an aggressive type of cancer with poor prognosis, short survival rate, and high mortality. Drug resistance is a major cause of treatment failure in the disease. MiR-331-3p has been reported to play an important role in several cancers. We previously showed that miR-331-3p is upregulated in pancreatic cancer and promotes pancreatic cancer cell proliferation and epithelial-to-mesenchymal transition–mediated metastasis by targeting ST7L. However, it is uncertain whether miR-331-3p is involved in drug resistance. Methods: We investigated the relationship between miR-331-3p and pancreatic cancer drug resistance. As part of this, microRNA mimics or inhibitors were transfected into pancreatic cancer cells. Quantitative polymerase chain reaction was used to detect miR-331-3p expression, and flow cytometry was used to detect cell apoptosis. The Cell Counting Kit-8 assay was used to measure the IC50 values of gemcitabine in pancreatic cancer cells. The expression of multidrug resistance protein 1, multidrug resistance-related protein 1, breast cancer resistance protein, β-Catenin, c-Myc, Cyclin D1, Bcl-2, and Caspase-3 was evaluated by Western blotting. Results: We confirmed that miR-331-3p is upregulated in gemcitabine-treated pancreatic cancer cells and plasma from chemotherapy patients. We also confirmed that miR-331-3p inhibition decreased drug resistance by regulating cell apoptosis and multidrug resistance protein 1, multidrug resistance-related protein 1, and breast cancer resistance protein expression in pancreatic cancer cells, whereas miR-331-3p overexpression had the opposite effect. We further demonstrated that miR-331-3p effects in drug resistance were partially reversed by ST7L overexpression. In addition, overexpression of miR-331-3p activated Wnt/β-catenin signaling in pancreatic cancer cells, and ST7L overexpression restored activation of Wnt/β-catenin signaling. Conclusions: Taken together, our data demonstrate that miR-331-3p contributes to drug resistance by activating Wnt/β-catenin signaling via ST7L in pancreatic cancer cells. These data provide a theoretical basis for new targeted therapies in the future.


Sign in / Sign up

Export Citation Format

Share Document