Improved Antitumor Activity and Tumor Targeting of NH2-Terminal-Specific PEGylated Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand

Authors
Chae, Su YoungKim, Tae HyungPark, KyeongsoonJin, Cheng-HaoSon, SoheeLee, SeulkiYoun, Yu SeokKim, KwangmeyungJo, Dong-GyuKwon, Ick ChanChen, XiaoyuanLee, Kang Choon
Issue Date
2010-06
Publisher
AMER ASSOC CANCER RESEARCH
Citation
MOLECULAR CANCER THERAPEUTICS, v.9, no.6, pp.1719 - 1729
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered an attractive anticancer agent due to its tumor cell-specific cytotoxicity. However, its low stability, solubility, unexpected side effects, and weak pharmacokinetic profiles restrict its successful clinical application. To develop efficient TRAIL-based anticancer biotherapeutics, a new version of trimeric TRAIL was constructed by incorporating trimer-forming zipper sequences (HZ-TRAIL), and then NH2-terminal-specific PEGylation was done to produce PEGylated TRAIL (PEG-HZ-TRAIL). The biological, physicochemical, and pharmaceutical characteristics of PEG-HZ-TRAIL were then investigated using various in vitro and in vivo experiments, including a cell-based cytotoxicity test, a solubility test, pharmacokinetic analysis, and antitumor efficacy evaluations. Although slight activity loss occurred after PEGylation, PEG-HZ-TRAIL showed excellent tumor cell-specific cytotoxic effects via apoptotic pathways with negligible normal cell toxicity. The stability and pharmacokinetic problems of HZ-TRAIL were successfully overcome by PEGylation. Furthermore, in vivo antitumor tests revealed that PEG-HZ-TRAIL treatment enhanced therapeutic potentials compared with HZ-TRAIL in tumor xenograft animal models, and these enhancements were attributed to its better pharmacokinetic properties and tumor-targeting performance. These findings show that PEG-HZ-TRAIL administration provides an effective antitumor treatment, which exhibits superior tumor targeting and better inhibits tumor growth, and suggest that PEG-HZ-TRAIL should be considered a potential candidate for antitumor biotherapy. Mol Cancer Ther; 9(6); 1719-29. (C)2010 AACR.
Keywords
SITE-SPECIFIC PEGYLATION; TUMORICIDAL ACTIVITY; RECEPTOR AGONISTS; CANCER-CELLS; TNF-ALPHA; IN-VIVO; TRAIL; DEATH; DELIVERY; PHARMACOKINETICS; SITE-SPECIFIC PEGYLATION; TUMORICIDAL ACTIVITY; RECEPTOR AGONISTS; CANCER-CELLS; TNF-ALPHA; IN-VIVO; TRAIL; DEATH; DELIVERY; PHARMACOKINETICS
ISSN
1535-7163
URI
https://pubs.kist.re.kr/handle/201004/131418
DOI
10.1158/1535-7163.MCT-09-1076
Appears in Collections:
KIST Article > 2010
Files in This Item:
There are no files associated with this item.
Export
RIS (EndNote)
XLS (Excel)
XML

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE