Please use this identifier to cite or link to this item: http://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/1583
Title: Supramolecular Nanoparticles That Target Phosphoinositide-3-Kinase Overcome Insulin Resistance and Exert Pronounced Antitumor Efficacy
Authors: Kulkarni, Ashish A.
Roy, Bhaskar
Rao, Poornima S.
Wyant, Gregory A.
Mahmoud, Ayaat
Ramachandran, Madhumitha
Sengupta, Poulomi
Goldman, Aaron
Kotamraju, Venkata Ramana
BASU, SUDIPTA
Mashelkar, Raghunath A.
Ruoslahti, Erkki
Dinulescu, Daniela M.
Sengupta, Shiladitya
Dept. of Chemistry
Keywords: World Health Organization
PI3K family
Nanovectors capitalize
Dichloromethane
Cholesterol conjugate
2013
Issue Date: Dec-2013
Publisher: American Association for Cancer Research
Citation: Cancer Research, 73 (23), 6987-6997.
Abstract: The centrality of phosphoinositide-3-kinase (PI3K) in cancer etiology is well established, but clinical translation of PI3K inhibitors has been limited by feedback signaling, suboptimal intratumoral concentration, and an insulin resistance “class effect.” This study was designed to explore the use of supramolecular nanochemistry for targeting PI3K to enhance antitumor efficacy and potentially overcome these limitations. PI3K inhibitor structures were rationally modified using a cholesterol-based derivative, facilitating supramolecular nanoassembly with L-α-phosphatidylcholine and DSPE-PEG [1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polythylene glycol)]. The supramolecular nanoparticles (SNP) that were assembled were physicochemically characterized and functionally evaluated in vitro. Antitumor efficacy was quantified in vivo using 4T1 breast cancer and K-RasLSL/+/Ptenfl/fl ovarian cancer models, with effects on glucose homeostasis evaluated using an insulin sensitivity test. The use of PI103 and PI828 as surrogate molecules to engineer the SNPs highlighted the need to keep design principles in perspective; specifically, potency of the active molecule and the linker chemistry were critical principles for efficacy, similar to antibody–drug conjugates. We found that the SNPs exerted a temporally sustained inhibition of phosphorylation of Akt, mTOR, S6K, and 4EBP in vivo. These effects were associated with increased antitumor efficacy and survival as compared with PI103 and PI828. Efficacy was further increased by decorating the nanoparticle surface with tumor-homing peptides. Notably, the use of SNPs abrogated the insulin resistance that has been associated widely with other PI3K inhibitors. This study provides a preclinical foundation for the use of supramolecular nanochemistry to overcome current challenges associated with PI3K inhibitors, offering a paradigm for extension to other molecularly targeted therapeutics being explored for cancer treatment.
URI: http://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/1583
https://doi.org/10.1158/0008-5472.CAN-12-4477
ISSN: Aug-72
1538-7445
Appears in Collections:JOURNAL ARTICLES

Files in This Item:
There are no files associated with this item.


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