Cargando…

All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro

PURPOSE: Melanoma, which is initiated from melanocytes, is the most fatal type of skin cancer. Melanoma-initiating cells significantly contribute to the initiation, metastasis, and recurrence of melanoma, and CD20 is a marker of melanoma-initiating cells. All-trans retinoic acid (ATRA) has been demo...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Xingyu, Zhang, Zhiyuan, Yang, Shengfeng, Chen, Hairong, Wang, Dan, Li, Jun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove Medical Press 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6163018/
https://www.ncbi.nlm.nih.gov/pubmed/30288053
http://dx.doi.org/10.2147/OTT.S169957
_version_ 1783359274836033536
author Chen, Xingyu
Zhang, Zhiyuan
Yang, Shengfeng
Chen, Hairong
Wang, Dan
Li, Jun
author_facet Chen, Xingyu
Zhang, Zhiyuan
Yang, Shengfeng
Chen, Hairong
Wang, Dan
Li, Jun
author_sort Chen, Xingyu
collection PubMed
description PURPOSE: Melanoma, which is initiated from melanocytes, is the most fatal type of skin cancer. Melanoma-initiating cells significantly contribute to the initiation, metastasis, and recurrence of melanoma, and CD20 is a marker of melanoma-initiating cells. All-trans retinoic acid (ATRA) has been demonstrated to induce differentiation, inhibit proliferation, and promote the apoptosis of cancer cells and cancer-initiating cells (CICs). However, there has been no report on ATRA activity against melanoma-initiating cells. In this study, we examined the activity of ATRA against melanoma-initiating cells and developed ATRA-encapsulated poly(lactic-co-glycolic acid) (PLGA) nanoparticles, which were conjugated with a CD20 antibody (ATRA-PNP-CD20) for targeted delivery of ATRA to CD20(+) melanoma-initiating cells. MATERIALS AND METHODS: The effects of ATRA and ATRA-PNP-CD20 against melanoma-initiating cells were investigated using a cytotoxicity assay, tumorsphere formation assay, and flow cytometry. RESULTS: ATRA-PNP-CD20 had a size of 126.9 nm and a negative zeta potential. The drug-loading capacity of ATRA-PNP-CD20 was 8.7%, and ATRA-PNP-CD20 displayed a sustained release of ATRA for 144 hours. The results showed that ATRA-PNP-CD20 could effectively and specifically deliver ATRA to CD20(+) melanoma-initiating cells, achieving superior inhibitory effects against CD20(+) melanoma-initiating cells compared with those of free ATRA and nontargeted nanoparticles. To the best of our knowledge, we report for the first time a potent activity of ATRA against CD20(+) melanoma-initiating cells, targeted drug delivery of ATRA via nanoparticles to melanoma-initiating cells, and the achievement of a superior inhibitory effect against melanoma-initiating cells by using a CD20 antibody. CONCLUSION: ATRA-PNP-CD20 represents a promising tool for eliminating melanoma-initiating cells and shows a potential for the therapy of melanoma.
format Online
Article
Text
id pubmed-6163018
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Dove Medical Press
record_format MEDLINE/PubMed
spelling pubmed-61630182018-10-04 All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro Chen, Xingyu Zhang, Zhiyuan Yang, Shengfeng Chen, Hairong Wang, Dan Li, Jun Onco Targets Ther Original Research PURPOSE: Melanoma, which is initiated from melanocytes, is the most fatal type of skin cancer. Melanoma-initiating cells significantly contribute to the initiation, metastasis, and recurrence of melanoma, and CD20 is a marker of melanoma-initiating cells. All-trans retinoic acid (ATRA) has been demonstrated to induce differentiation, inhibit proliferation, and promote the apoptosis of cancer cells and cancer-initiating cells (CICs). However, there has been no report on ATRA activity against melanoma-initiating cells. In this study, we examined the activity of ATRA against melanoma-initiating cells and developed ATRA-encapsulated poly(lactic-co-glycolic acid) (PLGA) nanoparticles, which were conjugated with a CD20 antibody (ATRA-PNP-CD20) for targeted delivery of ATRA to CD20(+) melanoma-initiating cells. MATERIALS AND METHODS: The effects of ATRA and ATRA-PNP-CD20 against melanoma-initiating cells were investigated using a cytotoxicity assay, tumorsphere formation assay, and flow cytometry. RESULTS: ATRA-PNP-CD20 had a size of 126.9 nm and a negative zeta potential. The drug-loading capacity of ATRA-PNP-CD20 was 8.7%, and ATRA-PNP-CD20 displayed a sustained release of ATRA for 144 hours. The results showed that ATRA-PNP-CD20 could effectively and specifically deliver ATRA to CD20(+) melanoma-initiating cells, achieving superior inhibitory effects against CD20(+) melanoma-initiating cells compared with those of free ATRA and nontargeted nanoparticles. To the best of our knowledge, we report for the first time a potent activity of ATRA against CD20(+) melanoma-initiating cells, targeted drug delivery of ATRA via nanoparticles to melanoma-initiating cells, and the achievement of a superior inhibitory effect against melanoma-initiating cells by using a CD20 antibody. CONCLUSION: ATRA-PNP-CD20 represents a promising tool for eliminating melanoma-initiating cells and shows a potential for the therapy of melanoma. Dove Medical Press 2018-09-25 /pmc/articles/PMC6163018/ /pubmed/30288053 http://dx.doi.org/10.2147/OTT.S169957 Text en © 2018 Chen et al. This work is published and licensed by Dove Medical Press Limited The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed.
spellingShingle Original Research
Chen, Xingyu
Zhang, Zhiyuan
Yang, Shengfeng
Chen, Hairong
Wang, Dan
Li, Jun
All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
title All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
title_full All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
title_fullStr All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
title_full_unstemmed All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
title_short All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
title_sort all-trans retinoic acid-encapsulated, cd20 antibody-conjugated poly(lactic-co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6163018/
https://www.ncbi.nlm.nih.gov/pubmed/30288053
http://dx.doi.org/10.2147/OTT.S169957
work_keys_str_mv AT chenxingyu alltransretinoicacidencapsulatedcd20antibodyconjugatedpolylacticcoglycolicacidnanoparticleseffectivelytargetandeliminatemelanomainitiatingcellsinvitro
AT zhangzhiyuan alltransretinoicacidencapsulatedcd20antibodyconjugatedpolylacticcoglycolicacidnanoparticleseffectivelytargetandeliminatemelanomainitiatingcellsinvitro
AT yangshengfeng alltransretinoicacidencapsulatedcd20antibodyconjugatedpolylacticcoglycolicacidnanoparticleseffectivelytargetandeliminatemelanomainitiatingcellsinvitro
AT chenhairong alltransretinoicacidencapsulatedcd20antibodyconjugatedpolylacticcoglycolicacidnanoparticleseffectivelytargetandeliminatemelanomainitiatingcellsinvitro
AT wangdan alltransretinoicacidencapsulatedcd20antibodyconjugatedpolylacticcoglycolicacidnanoparticleseffectivelytargetandeliminatemelanomainitiatingcellsinvitro
AT lijun alltransretinoicacidencapsulatedcd20antibodyconjugatedpolylacticcoglycolicacidnanoparticleseffectivelytargetandeliminatemelanomainitiatingcellsinvitro