Cargando…

Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model

BACKGROUND: The poor prognosis of patients with drug resistant ovarian cancer and the lack of targeted therapy have raised the need for alternative treatments. Albendazole (ABZ) is an anti-parasite compound capable of impairing microtubule formation. We hypothesized that ABZ could be repurposed as a...

Descripción completa

Detalles Bibliográficos
Autores principales: Noorani, Lubna, Stenzel, Martina, Liang, Roger, Pourgholami, Mohammad H, Morris, David L
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4409778/
https://www.ncbi.nlm.nih.gov/pubmed/25890381
http://dx.doi.org/10.1186/s12951-015-0082-8
_version_ 1782368236012044288
author Noorani, Lubna
Stenzel, Martina
Liang, Roger
Pourgholami, Mohammad H
Morris, David L
author_facet Noorani, Lubna
Stenzel, Martina
Liang, Roger
Pourgholami, Mohammad H
Morris, David L
author_sort Noorani, Lubna
collection PubMed
description BACKGROUND: The poor prognosis of patients with drug resistant ovarian cancer and the lack of targeted therapy have raised the need for alternative treatments. Albendazole (ABZ) is an anti-parasite compound capable of impairing microtubule formation. We hypothesized that ABZ could be repurposed as a potential anti-angiogenic drug due to its potent inhibition of vascular endothelial growth factor (VEGF) in ovarian cancer with ascites. However, the poor aqueous solubility of ABZ limits its potential for cancer therapy. In this study, we have assembled ABZ with bovine serum albumin into nanoparticles with a size range of 7–10 nm (BSA-ABZ) and 200–250 nm (Nab-ABZ). We further examined the anticancer effects of ABZ carrying nanoparticles in ovarian cancer cells, in both in vitro and in vivo models. RESULTS: Drug release studies demonstrated that about 93% of ABZ was released from BSA-ABZ 10 nm in comparison to 83% from Nab-ABZ 200 nm at pH 7.4 in 8 days. In vitro cell proliferation studies showed that the BSA-ABZ 10 nm exhibited the highest killing efficacy of ovarian cancer cells with surprisingly least toxicity to healthy ovarian epithelial cells. Confocal microscopy and fluorescence activated cell sorting analysis (FACS) revealed more efficient internalization of the BSA-ABZ 10 nm by cancer cells. For in vivo studies, we examined the tumor growth, ascites formation and the expression of VEGF and secreted protein acidic and rich in cysteine (SPARC) in tumor samples and only VEGF in plasma samples. The BSA-ABZ 10 nm reduced the tumor burden significantly (p < 0.02) at a much lower drug dose (10 μg/ml) compare to free drug. Both formulations were capable of suppressing the ascites volume significantly (p < 0.05) and reducing the number of ascites cells. The expression of VEGF and SPARC was also reduced, which indicates the underlying therapeutic mechanism of the ABZ. CONCLUSION: Our data suggest that the BSA-ABZ may hold promise for the treatment and control of progression of ovarian cancer with ascites. However further studies are required to examine the efficacy of both the formulations in aggressive models of recurrent ovarian cancer with respect to particle size and dosing parameters. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12951-015-0082-8) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4409778
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-44097782015-04-26 Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model Noorani, Lubna Stenzel, Martina Liang, Roger Pourgholami, Mohammad H Morris, David L J Nanobiotechnology Research BACKGROUND: The poor prognosis of patients with drug resistant ovarian cancer and the lack of targeted therapy have raised the need for alternative treatments. Albendazole (ABZ) is an anti-parasite compound capable of impairing microtubule formation. We hypothesized that ABZ could be repurposed as a potential anti-angiogenic drug due to its potent inhibition of vascular endothelial growth factor (VEGF) in ovarian cancer with ascites. However, the poor aqueous solubility of ABZ limits its potential for cancer therapy. In this study, we have assembled ABZ with bovine serum albumin into nanoparticles with a size range of 7–10 nm (BSA-ABZ) and 200–250 nm (Nab-ABZ). We further examined the anticancer effects of ABZ carrying nanoparticles in ovarian cancer cells, in both in vitro and in vivo models. RESULTS: Drug release studies demonstrated that about 93% of ABZ was released from BSA-ABZ 10 nm in comparison to 83% from Nab-ABZ 200 nm at pH 7.4 in 8 days. In vitro cell proliferation studies showed that the BSA-ABZ 10 nm exhibited the highest killing efficacy of ovarian cancer cells with surprisingly least toxicity to healthy ovarian epithelial cells. Confocal microscopy and fluorescence activated cell sorting analysis (FACS) revealed more efficient internalization of the BSA-ABZ 10 nm by cancer cells. For in vivo studies, we examined the tumor growth, ascites formation and the expression of VEGF and secreted protein acidic and rich in cysteine (SPARC) in tumor samples and only VEGF in plasma samples. The BSA-ABZ 10 nm reduced the tumor burden significantly (p < 0.02) at a much lower drug dose (10 μg/ml) compare to free drug. Both formulations were capable of suppressing the ascites volume significantly (p < 0.05) and reducing the number of ascites cells. The expression of VEGF and SPARC was also reduced, which indicates the underlying therapeutic mechanism of the ABZ. CONCLUSION: Our data suggest that the BSA-ABZ may hold promise for the treatment and control of progression of ovarian cancer with ascites. However further studies are required to examine the efficacy of both the formulations in aggressive models of recurrent ovarian cancer with respect to particle size and dosing parameters. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12951-015-0082-8) contains supplementary material, which is available to authorized users. BioMed Central 2015-03-25 /pmc/articles/PMC4409778/ /pubmed/25890381 http://dx.doi.org/10.1186/s12951-015-0082-8 Text en © Noorani et al.; licensee BioMed Central. 2015 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Noorani, Lubna
Stenzel, Martina
Liang, Roger
Pourgholami, Mohammad H
Morris, David L
Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
title Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
title_full Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
title_fullStr Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
title_full_unstemmed Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
title_short Albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
title_sort albumin nanoparticles increase the anticancer efficacy of albendazole in ovarian cancer xenograft model
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4409778/
https://www.ncbi.nlm.nih.gov/pubmed/25890381
http://dx.doi.org/10.1186/s12951-015-0082-8
work_keys_str_mv AT nooranilubna albuminnanoparticlesincreasetheanticancerefficacyofalbendazoleinovariancancerxenograftmodel
AT stenzelmartina albuminnanoparticlesincreasetheanticancerefficacyofalbendazoleinovariancancerxenograftmodel
AT liangroger albuminnanoparticlesincreasetheanticancerefficacyofalbendazoleinovariancancerxenograftmodel
AT pourgholamimohammadh albuminnanoparticlesincreasetheanticancerefficacyofalbendazoleinovariancancerxenograftmodel
AT morrisdavidl albuminnanoparticlesincreasetheanticancerefficacyofalbendazoleinovariancancerxenograftmodel