Digital Repository

Supramolecular Nanoparticles That Target Phosphoinositide-3-Kinase Overcome Insulin Resistance and Exert Pronounced Antitumor Efficacy

Show simple item record

dc.contributor.author Kulkarni, Ashish A. en_US
dc.contributor.author Roy, Bhaskar en_US
dc.contributor.author Rao, Poornima S. en_US
dc.contributor.author Wyant, Gregory A. en_US
dc.contributor.author Mahmoud, Ayaat en_US
dc.contributor.author Ramachandran, Madhumitha en_US
dc.contributor.author Sengupta, Poulomi en_US
dc.contributor.author Goldman, Aaron en_US
dc.contributor.author Kotamraju, Venkata Ramana en_US
dc.contributor.author BASU, SUDIPTA en_US
dc.contributor.author Mashelkar, Raghunath A. en_US
dc.contributor.author Ruoslahti, Erkki en_US
dc.contributor.author Dinulescu, Daniela M. en_US
dc.contributor.author Sengupta, Shiladitya en_US
dc.date.accessioned 2019-02-14T05:00:09Z
dc.date.available 2019-02-14T05:00:09Z
dc.date.issued 2013-12 en_US
dc.identifier.citation Cancer Research, 73 (23), 6987-6997. en_US
dc.identifier.issn Aug-72 en_US
dc.identifier.issn 1538-7445 en_US
dc.identifier.uri http://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/1583
dc.identifier.uri https://doi.org/10.1158/0008-5472.CAN-12-4477 en_US
dc.description.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. en_US
dc.language.iso en en_US
dc.publisher American Association for Cancer Research en_US
dc.subject World Health Organization en_US
dc.subject PI3K family en_US
dc.subject Nanovectors capitalize en_US
dc.subject Dichloromethane en_US
dc.subject Cholesterol conjugate en_US
dc.subject 2013 en_US
dc.title Supramolecular Nanoparticles That Target Phosphoinositide-3-Kinase Overcome Insulin Resistance and Exert Pronounced Antitumor Efficacy en_US
dc.type Article en_US
dc.contributor.department Dept. of Chemistry en_US
dc.identifier.sourcetitle Cancer Research en_US
dc.publication.originofpublisher Foreign en_US


Files in this item

Files Size Format View

There are no files associated with this item.

This item appears in the following Collection(s)

Show simple item record

Search Repository


Advanced Search

Browse

My Account