scholarly journals Increasing the potential of cell-penetrating peptides for cancer therapy using a new pentagonal scaffold

2019 ◽  
Vol 860 ◽  
pp. 172554 ◽  
Author(s):  
Diana Duarte ◽  
Alexandra G. Fraga ◽  
Jorge Pedrosa ◽  
Fátima Martel ◽  
Nuno Vale
2017 ◽  
Vol 53 (83) ◽  
pp. 11433-11436 ◽  
Author(s):  
Qilin Yu ◽  
Bing Zhang ◽  
Jianrong Li ◽  
Mingchun Li

Graphene oxide (GO) nanosheets grafted with actin targeting and cell penetrating peptides were designed.


2018 ◽  
Vol 6 (46) ◽  
pp. 7674-7683 ◽  
Author(s):  
Yanxi Liu ◽  
Qiuyi Li ◽  
Xiaofeng Xiong ◽  
Yuan Huang ◽  
Zhou Zhou

Targeting drugs at mitochondria may provide an effective means of inducing cell death for cancer therapy.


Author(s):  
Xiao Fu ◽  
Guiqiang Zhang ◽  
Yulin Zhang ◽  
Haifeng Sun ◽  
Shuang Yang ◽  
...  

Drug Delivery ◽  
2016 ◽  
Vol 23 (9) ◽  
pp. 3436-3443 ◽  
Author(s):  
Wen Lin ◽  
Xiangyang Xie ◽  
Yanfang Yang ◽  
Xudong Fu ◽  
Hong Liu ◽  
...  

Pharmaceutics ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 365
Author(s):  
Dina V. Hingorani ◽  
Maria F. Camargo ◽  
Maryam A. Quraishi ◽  
Stephen R. Adams ◽  
Sunil J. Advani

Recent advances in immunotherapy have revolutionized cancer therapy. Immunotherapies can engage the adaptive and innate arms of the immune system. Therapeutics targeting immune checkpoint inhibitors (i.e., CTLA-4; PD-1, and PD-L1) have shown efficacy for subsets of cancer patients by unleashing an adaptive antitumor immune response. Alternatively, small molecule immune modulators of the innate immune system such as toll-like receptor (TLR) agonists are being developed for cancer therapy. TLRs function as pattern recognition receptors to microbial products and are also involved in carcinogenesis. Reisquimod is a TLR 7/8 agonist that has antitumor efficacy. However, systemic delivery free resiquimod has proven to be challenging due to toxicity of nonspecific TLR 7/8 activation. Therefore, we developed a targeted peptide-drug conjugate strategy for systemic delivery of resiquimod. We designed an activatable cell penetrating peptide to deliver resiquimod specifically to the tumor tissue while avoiding normal tissues. The activatable cell penetrating peptide (ACPP) scaffold undergoes enzymatic cleavage by matrix metalloproteinases 2/9 in the extracellular matrix followed by intracellular lysosomal cathepsin B mediated release of the free resiquimod. Importantly, when conjugated to ACPP; the tumor tissue concentration of resiquimod was more than 1000-fold greater than that of surrounding non-cancerous tissue. Moreover, systemic ACPP-resiquimod delivery produced comparable therapeutic efficacy to localized free resiquimod in syngeneic murine tumors. These results highlight a precision peptide-drug conjugate delivery.


Sign in / Sign up

Export Citation Format

Share Document