Cargando…

Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial

BACKGROUND: Cancer metastasis caused by circulating tumor cells (CTCs) accounts for 90% cancer-related death worldwide. Blocking the circulation of CTCs in bloodstream and their hetero-adhesion to vascular endothelia of the distant metastatic organs may prevent cancer metastasis. Nanomaterial-based...

Descripción completa

Detalles Bibliográficos
Autores principales: Xie, Jingjing, Dong, Haiyan, Chen, Hongning, Zhao, Rongli, Sinko, Patrick J, Shen, Weiyu, Wang, Jichuang, Lu, Yusheng, Yang, Xiang, Xie, Fangwei, Jia, Lee
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4320453/
https://www.ncbi.nlm.nih.gov/pubmed/25643843
http://dx.doi.org/10.1186/s12951-015-0072-x
_version_ 1782356116573782016
author Xie, Jingjing
Dong, Haiyan
Chen, Hongning
Zhao, Rongli
Sinko, Patrick J
Shen, Weiyu
Wang, Jichuang
Lu, Yusheng
Yang, Xiang
Xie, Fangwei
Jia, Lee
author_facet Xie, Jingjing
Dong, Haiyan
Chen, Hongning
Zhao, Rongli
Sinko, Patrick J
Shen, Weiyu
Wang, Jichuang
Lu, Yusheng
Yang, Xiang
Xie, Fangwei
Jia, Lee
author_sort Xie, Jingjing
collection PubMed
description BACKGROUND: Cancer metastasis caused by circulating tumor cells (CTCs) accounts for 90% cancer-related death worldwide. Blocking the circulation of CTCs in bloodstream and their hetero-adhesion to vascular endothelia of the distant metastatic organs may prevent cancer metastasis. Nanomaterial-based intervention with adhesion between CTCs and endothelia has not been reported. Driven by the novel idea that multivalent conjugation of EpCAM and Slex antibodies to dendrimer surface may enhance the capacity and specificity of the nanomaterial conjugates for capturing and down-regulating colorectal CTCs, we conjugated the dendrimer nanomaterial with the EpCAM and Slex antibodies, and examined the capacity of the dual antibody-coated nanomaterial for their roles in interrupting CTCs-related cancer metastasis. RESULTS: The antibody-coated nanomaterial was synthesized and characterized. The conjugates specifically bound and captured colon cancer cells SW620. The conjugate inhibited the cells’ viability and their adhesion to fibronectin (Fn)-coated substrate or human umbilical vein endothelial cells (HUVECs) in a concentration-dependent manner. In comparison with SW480 and LoVo cell lines, the activity and adhesion of SW620 to Fn-coated substrate and HUVECs were more specifically inhibited by the dual antibody conjugate because of the higher levels of EpCAM and Slex on SW620 cell surface. The hetero-adhesion between SW620 and Fn-coated substrate, or HUVECs was inhibited by about 60-70%. The dual conjugate showed the inhibition capacity more significant than its corresponding single antibody conjugates. CONCLUSIONS: The present study provides the new evidence that coating nanomaterials with more than one antibody against CTCs may effectively interfere with the interaction between SW620 and HUVECs. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12951-015-0072-x) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4320453
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-43204532015-02-08 Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial Xie, Jingjing Dong, Haiyan Chen, Hongning Zhao, Rongli Sinko, Patrick J Shen, Weiyu Wang, Jichuang Lu, Yusheng Yang, Xiang Xie, Fangwei Jia, Lee J Nanobiotechnology Research BACKGROUND: Cancer metastasis caused by circulating tumor cells (CTCs) accounts for 90% cancer-related death worldwide. Blocking the circulation of CTCs in bloodstream and their hetero-adhesion to vascular endothelia of the distant metastatic organs may prevent cancer metastasis. Nanomaterial-based intervention with adhesion between CTCs and endothelia has not been reported. Driven by the novel idea that multivalent conjugation of EpCAM and Slex antibodies to dendrimer surface may enhance the capacity and specificity of the nanomaterial conjugates for capturing and down-regulating colorectal CTCs, we conjugated the dendrimer nanomaterial with the EpCAM and Slex antibodies, and examined the capacity of the dual antibody-coated nanomaterial for their roles in interrupting CTCs-related cancer metastasis. RESULTS: The antibody-coated nanomaterial was synthesized and characterized. The conjugates specifically bound and captured colon cancer cells SW620. The conjugate inhibited the cells’ viability and their adhesion to fibronectin (Fn)-coated substrate or human umbilical vein endothelial cells (HUVECs) in a concentration-dependent manner. In comparison with SW480 and LoVo cell lines, the activity and adhesion of SW620 to Fn-coated substrate and HUVECs were more specifically inhibited by the dual antibody conjugate because of the higher levels of EpCAM and Slex on SW620 cell surface. The hetero-adhesion between SW620 and Fn-coated substrate, or HUVECs was inhibited by about 60-70%. The dual conjugate showed the inhibition capacity more significant than its corresponding single antibody conjugates. CONCLUSIONS: The present study provides the new evidence that coating nanomaterials with more than one antibody against CTCs may effectively interfere with the interaction between SW620 and HUVECs. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12951-015-0072-x) contains supplementary material, which is available to authorized users. BioMed Central 2015-02-03 /pmc/articles/PMC4320453/ /pubmed/25643843 http://dx.doi.org/10.1186/s12951-015-0072-x Text en © Xie 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
Xie, Jingjing
Dong, Haiyan
Chen, Hongning
Zhao, Rongli
Sinko, Patrick J
Shen, Weiyu
Wang, Jichuang
Lu, Yusheng
Yang, Xiang
Xie, Fangwei
Jia, Lee
Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
title Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
title_full Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
title_fullStr Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
title_full_unstemmed Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
title_short Exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
title_sort exploring cancer metastasis prevention strategy: interrupting adhesion of cancer cells to vascular endothelia of potential metastatic tissues by antibody-coated nanomaterial
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4320453/
https://www.ncbi.nlm.nih.gov/pubmed/25643843
http://dx.doi.org/10.1186/s12951-015-0072-x
work_keys_str_mv AT xiejingjing exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT donghaiyan exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT chenhongning exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT zhaorongli exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT sinkopatrickj exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT shenweiyu exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT wangjichuang exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT luyusheng exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT yangxiang exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT xiefangwei exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial
AT jialee exploringcancermetastasispreventionstrategyinterruptingadhesionofcancercellstovascularendotheliaofpotentialmetastatictissuesbyantibodycoatednanomaterial