Cargando…

A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer

Of patients with castrate resistant prostate cancer (CRPC), less than 25–33% survive more than five years. Recent studies have implicated estrogen, acting either alone or synergistically with androgens in the development of castrate resistant prostate cancer. Several in vitro and in vivo studies, as...

Descripción completa

Detalles Bibliográficos
Autores principales: Taurin, Sebastien, Nehoff, Hayley, van Aswegen, Thalita, Rosengren, Rhonda J., Greish, Khaled
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi Publishing Corporation 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3932713/
https://www.ncbi.nlm.nih.gov/pubmed/24689036
http://dx.doi.org/10.1155/2014/323594
_version_ 1782304825840500736
author Taurin, Sebastien
Nehoff, Hayley
van Aswegen, Thalita
Rosengren, Rhonda J.
Greish, Khaled
author_facet Taurin, Sebastien
Nehoff, Hayley
van Aswegen, Thalita
Rosengren, Rhonda J.
Greish, Khaled
author_sort Taurin, Sebastien
collection PubMed
description Of patients with castrate resistant prostate cancer (CRPC), less than 25–33% survive more than five years. Recent studies have implicated estrogen, acting either alone or synergistically with androgens in the development of castrate resistant prostate cancer. Several in vitro and in vivo studies, as well as a limited number of clinical trials, have highlighted the potential of selective estrogen receptor modulators, such as raloxifene (Ral) for the treatment of castrate resistant prostate cancer. However, the poor oral bioavailability and metabolism of selective estrogen receptor modulators limit their efficiency in clinical application. To overcome these limitations, we have used styrene co-maleic acid (SMA) micelle to encapsulate raloxifene. Compared to free drug, SMA-Ral micelles had 132 and 140% higher cytotoxicity against PC3 and DU 145 prostate cell lines, respectively. SMA-Ral effectively inhibits cell cycle progression, increases apoptosis, and alters the integrity of tumor spheroid models. In addition, the micellar system induced changes in expression and localization of estrogen receptors, epidermal growth factor receptor (EGFR), and downstream effectors associated with cell proliferation and survival. Finally, SMA-Ral treatment decreased migration and invasion of castrate resistant prostate cancer cell lines. In conclusion, SMA-Ral micelles can potentially benefit new strategies for clinical management of castrate resistant prostate cancer.
format Online
Article
Text
id pubmed-3932713
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Hindawi Publishing Corporation
record_format MEDLINE/PubMed
spelling pubmed-39327132014-03-31 A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer Taurin, Sebastien Nehoff, Hayley van Aswegen, Thalita Rosengren, Rhonda J. Greish, Khaled Biomed Res Int Research Article Of patients with castrate resistant prostate cancer (CRPC), less than 25–33% survive more than five years. Recent studies have implicated estrogen, acting either alone or synergistically with androgens in the development of castrate resistant prostate cancer. Several in vitro and in vivo studies, as well as a limited number of clinical trials, have highlighted the potential of selective estrogen receptor modulators, such as raloxifene (Ral) for the treatment of castrate resistant prostate cancer. However, the poor oral bioavailability and metabolism of selective estrogen receptor modulators limit their efficiency in clinical application. To overcome these limitations, we have used styrene co-maleic acid (SMA) micelle to encapsulate raloxifene. Compared to free drug, SMA-Ral micelles had 132 and 140% higher cytotoxicity against PC3 and DU 145 prostate cell lines, respectively. SMA-Ral effectively inhibits cell cycle progression, increases apoptosis, and alters the integrity of tumor spheroid models. In addition, the micellar system induced changes in expression and localization of estrogen receptors, epidermal growth factor receptor (EGFR), and downstream effectors associated with cell proliferation and survival. Finally, SMA-Ral treatment decreased migration and invasion of castrate resistant prostate cancer cell lines. In conclusion, SMA-Ral micelles can potentially benefit new strategies for clinical management of castrate resistant prostate cancer. Hindawi Publishing Corporation 2014 2014-02-06 /pmc/articles/PMC3932713/ /pubmed/24689036 http://dx.doi.org/10.1155/2014/323594 Text en Copyright © 2014 Sebastien Taurin et al. https://creativecommons.org/licenses/by/3.0/ This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Taurin, Sebastien
Nehoff, Hayley
van Aswegen, Thalita
Rosengren, Rhonda J.
Greish, Khaled
A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer
title A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer
title_full A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer
title_fullStr A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer
title_full_unstemmed A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer
title_short A Novel Role for Raloxifene Nanomicelles in Management of Castrate Resistant Prostate Cancer
title_sort novel role for raloxifene nanomicelles in management of castrate resistant prostate cancer
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3932713/
https://www.ncbi.nlm.nih.gov/pubmed/24689036
http://dx.doi.org/10.1155/2014/323594
work_keys_str_mv AT taurinsebastien anovelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT nehoffhayley anovelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT vanaswegenthalita anovelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT rosengrenrhondaj anovelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT greishkhaled anovelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT taurinsebastien novelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT nehoffhayley novelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT vanaswegenthalita novelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT rosengrenrhondaj novelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer
AT greishkhaled novelroleforraloxifenenanomicellesinmanagementofcastrateresistantprostatecancer