Cargando…

Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro

BACKGROUND: Aptamer-based tumor targeted drug delivery system is a promising approach that may increase the efficacy of chemotherapy and reduce the related toxicity. HER2 protein is an attractive target for tumor-specific drug delivery because of its overexpression in multiple malignancies, includin...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Zhe, Duan, Jin-Hong, Song, Yong-Mei, Ma, Jie, Wang, Feng-Dan, Lu, Xin, Yang, Xian-Da
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3583217/
https://www.ncbi.nlm.nih.gov/pubmed/22817844
http://dx.doi.org/10.1186/1479-5876-10-148
_version_ 1782475407994388480
author Liu, Zhe
Duan, Jin-Hong
Song, Yong-Mei
Ma, Jie
Wang, Feng-Dan
Lu, Xin
Yang, Xian-Da
author_facet Liu, Zhe
Duan, Jin-Hong
Song, Yong-Mei
Ma, Jie
Wang, Feng-Dan
Lu, Xin
Yang, Xian-Da
author_sort Liu, Zhe
collection PubMed
description BACKGROUND: Aptamer-based tumor targeted drug delivery system is a promising approach that may increase the efficacy of chemotherapy and reduce the related toxicity. HER2 protein is an attractive target for tumor-specific drug delivery because of its overexpression in multiple malignancies, including breast, gastric, ovarian, and lung cancers. METHODS: In this paper, we developed a new HER2 aptamer (HB5) by using systematic evolution of ligands by exponential enrichment technology (SELEX) and exploited its role as a targeting ligand for delivering doxorubicin (Dox) to breast cancer cells in vitro. RESULTS: The selected aptamer was an 86-nucleotide DNA molecule that bound to an epitope peptide of HER2 with a K(d) of 18.9 nM. The aptamer also bound to the extracellular domain (ECD) of HER2 protein with a K(d)of 316 nM, and had minimal cross reactivity to albumin or trypsin. In addition, the aptamer was found to preferentially bind to HER2-positive but not HER2-negative breast cancer cells. An aptamer-doxorubicin complex (Apt-Dox) was formulated by intercalating Dox into the DNA structure of HB5. The Apt-Dox complex could selectively deliver Dox to HER2-positive breast cancer cells while reducing the drug intake by HER2-negative cells in vitro. Moreover, Apt-Dox retained the cytotoxicity of Dox against HER2-positive breast cancer cells, but reduced the cytotoxicity to HER2-negative cells. CONCLUSIONS: The results suggest that the selected HER2 aptamer may have application potentials in targeted therapy against HER2-positive breast cancer cells.
format Online
Article
Text
id pubmed-3583217
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-35832172013-02-28 Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro Liu, Zhe Duan, Jin-Hong Song, Yong-Mei Ma, Jie Wang, Feng-Dan Lu, Xin Yang, Xian-Da J Transl Med Research BACKGROUND: Aptamer-based tumor targeted drug delivery system is a promising approach that may increase the efficacy of chemotherapy and reduce the related toxicity. HER2 protein is an attractive target for tumor-specific drug delivery because of its overexpression in multiple malignancies, including breast, gastric, ovarian, and lung cancers. METHODS: In this paper, we developed a new HER2 aptamer (HB5) by using systematic evolution of ligands by exponential enrichment technology (SELEX) and exploited its role as a targeting ligand for delivering doxorubicin (Dox) to breast cancer cells in vitro. RESULTS: The selected aptamer was an 86-nucleotide DNA molecule that bound to an epitope peptide of HER2 with a K(d) of 18.9 nM. The aptamer also bound to the extracellular domain (ECD) of HER2 protein with a K(d)of 316 nM, and had minimal cross reactivity to albumin or trypsin. In addition, the aptamer was found to preferentially bind to HER2-positive but not HER2-negative breast cancer cells. An aptamer-doxorubicin complex (Apt-Dox) was formulated by intercalating Dox into the DNA structure of HB5. The Apt-Dox complex could selectively deliver Dox to HER2-positive breast cancer cells while reducing the drug intake by HER2-negative cells in vitro. Moreover, Apt-Dox retained the cytotoxicity of Dox against HER2-positive breast cancer cells, but reduced the cytotoxicity to HER2-negative cells. CONCLUSIONS: The results suggest that the selected HER2 aptamer may have application potentials in targeted therapy against HER2-positive breast cancer cells. BioMed Central 2012-07-20 /pmc/articles/PMC3583217/ /pubmed/22817844 http://dx.doi.org/10.1186/1479-5876-10-148 Text en Copyright ©2012 Liu et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research
Liu, Zhe
Duan, Jin-Hong
Song, Yong-Mei
Ma, Jie
Wang, Feng-Dan
Lu, Xin
Yang, Xian-Da
Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro
title Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro
title_full Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro
title_fullStr Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro
title_full_unstemmed Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro
title_short Novel HER2 Aptamer Selectively Delivers Cytotoxic Drug to HER2-positive Breast Cancer Cells in Vitro
title_sort novel her2 aptamer selectively delivers cytotoxic drug to her2-positive breast cancer cells in vitro
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3583217/
https://www.ncbi.nlm.nih.gov/pubmed/22817844
http://dx.doi.org/10.1186/1479-5876-10-148
work_keys_str_mv AT liuzhe novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro
AT duanjinhong novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro
AT songyongmei novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro
AT majie novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro
AT wangfengdan novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro
AT luxin novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro
AT yangxianda novelher2aptamerselectivelydeliverscytotoxicdrugtoher2positivebreastcancercellsinvitro