Cargando…

Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma

Rationale: Tumor-associated fibroblasts (TAFs) play a critical role in the suppressive immune tumor microenvironment (TME), compromising the efficacy of immunotherapy. To overcome this therapeutic hurdle, we developed a nanoemulsion (NE) formulation to deliver fraxinellone (Frax), an anti-fibrotic m...

Descripción completa

Detalles Bibliográficos
Autores principales: Hou, Lin, Liu, Qi, Shen, Limei, Liu, Yun, Zhang, Xueqiong, Chen, Fengqian, Huang, Leaf
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6071534/
https://www.ncbi.nlm.nih.gov/pubmed/30083259
http://dx.doi.org/10.7150/thno.24821
_version_ 1783343887695216640
author Hou, Lin
Liu, Qi
Shen, Limei
Liu, Yun
Zhang, Xueqiong
Chen, Fengqian
Huang, Leaf
author_facet Hou, Lin
Liu, Qi
Shen, Limei
Liu, Yun
Zhang, Xueqiong
Chen, Fengqian
Huang, Leaf
author_sort Hou, Lin
collection PubMed
description Rationale: Tumor-associated fibroblasts (TAFs) play a critical role in the suppressive immune tumor microenvironment (TME), compromising the efficacy of immunotherapy. To overcome this therapeutic hurdle, we developed a nanoemulsion (NE) formulation to deliver fraxinellone (Frax), an anti-fibrotic medicine, to TAFs, as an approach to reverse immunosuppressive TME of desmoplastic melanoma. Methods: Frax NE was prepared by an ultrasonic emulsification method. The tumor inhibition effect was evaluated by immunofluorescence staining, masson trichrome staining and western blot analysis. Immune cell populations in tumor and LNs were detected by flow cytometry. Results: This Frax NE, with a particle size of around 145 nm, can efficiently accumulate in the tumor site after systemic administration and was taken up by TAFs and tumor cells. A significant decrease in TAFs and stroma deposition was observed after intravenous administration of Frax NE, and Frax NE treatment also remolded the tumor immune microenvironment, as was reflected by an increase of natural-killer cells, cytotoxic T cells (CTLs) as well as a decrease of regulatory B cells, and myeloid-derived suppressor cells in the TME. In addition, after treatment by Frax NEs, T helper 1 (Th1) cytokines of interferon gamma (IFN-γ), which effectively elicit anti-tumor immunity, were enhanced. Transforming growth factor-β (TGF-β), chemokine (C-C motif) ligand 2 (CCL2) and interleukin 6 (IL6), which inhibit the development of anti-tumor immunity, were reduced. Although Frax NE demonstrated an inhibitory effect on tumor growth, this mono-therapy could only achieve partial antitumor efficacy, and the tumor growth effect was not maintained long-term after dosing stopped. Therefore, a tumor-specific peptide vaccine was combined with Frax NEs. The combination led to enhanced tumor-specific T-cell infiltration, activated death receptors on the tumor cell surface, and induced increased apoptotic tumor cell death. Conclusion: Collectively, Frax NE combined with tumor-specific peptide vaccine might be an effective and safe strategy to remodel fibrotic TME, thereby enhancing immune response activation, resulting in a prolonged efficiency for advanced desmoplastic melanoma.
format Online
Article
Text
id pubmed-6071534
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-60715342018-08-06 Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma Hou, Lin Liu, Qi Shen, Limei Liu, Yun Zhang, Xueqiong Chen, Fengqian Huang, Leaf Theranostics Research Paper Rationale: Tumor-associated fibroblasts (TAFs) play a critical role in the suppressive immune tumor microenvironment (TME), compromising the efficacy of immunotherapy. To overcome this therapeutic hurdle, we developed a nanoemulsion (NE) formulation to deliver fraxinellone (Frax), an anti-fibrotic medicine, to TAFs, as an approach to reverse immunosuppressive TME of desmoplastic melanoma. Methods: Frax NE was prepared by an ultrasonic emulsification method. The tumor inhibition effect was evaluated by immunofluorescence staining, masson trichrome staining and western blot analysis. Immune cell populations in tumor and LNs were detected by flow cytometry. Results: This Frax NE, with a particle size of around 145 nm, can efficiently accumulate in the tumor site after systemic administration and was taken up by TAFs and tumor cells. A significant decrease in TAFs and stroma deposition was observed after intravenous administration of Frax NE, and Frax NE treatment also remolded the tumor immune microenvironment, as was reflected by an increase of natural-killer cells, cytotoxic T cells (CTLs) as well as a decrease of regulatory B cells, and myeloid-derived suppressor cells in the TME. In addition, after treatment by Frax NEs, T helper 1 (Th1) cytokines of interferon gamma (IFN-γ), which effectively elicit anti-tumor immunity, were enhanced. Transforming growth factor-β (TGF-β), chemokine (C-C motif) ligand 2 (CCL2) and interleukin 6 (IL6), which inhibit the development of anti-tumor immunity, were reduced. Although Frax NE demonstrated an inhibitory effect on tumor growth, this mono-therapy could only achieve partial antitumor efficacy, and the tumor growth effect was not maintained long-term after dosing stopped. Therefore, a tumor-specific peptide vaccine was combined with Frax NEs. The combination led to enhanced tumor-specific T-cell infiltration, activated death receptors on the tumor cell surface, and induced increased apoptotic tumor cell death. Conclusion: Collectively, Frax NE combined with tumor-specific peptide vaccine might be an effective and safe strategy to remodel fibrotic TME, thereby enhancing immune response activation, resulting in a prolonged efficiency for advanced desmoplastic melanoma. Ivyspring International Publisher 2018-06-13 /pmc/articles/PMC6071534/ /pubmed/30083259 http://dx.doi.org/10.7150/thno.24821 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Hou, Lin
Liu, Qi
Shen, Limei
Liu, Yun
Zhang, Xueqiong
Chen, Fengqian
Huang, Leaf
Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
title Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
title_full Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
title_fullStr Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
title_full_unstemmed Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
title_short Nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
title_sort nano-delivery of fraxinellone remodels tumor microenvironment and facilitates therapeutic vaccination in desmoplastic melanoma
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6071534/
https://www.ncbi.nlm.nih.gov/pubmed/30083259
http://dx.doi.org/10.7150/thno.24821
work_keys_str_mv AT houlin nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma
AT liuqi nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma
AT shenlimei nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma
AT liuyun nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma
AT zhangxueqiong nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma
AT chenfengqian nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma
AT huangleaf nanodeliveryoffraxinelloneremodelstumormicroenvironmentandfacilitatestherapeuticvaccinationindesmoplasticmelanoma