Cargando…

Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth

Therapeutic strategies aiming at mobilizing immune effector cells to kill tumor cells independent of tumor mutational load and MHC expression status are expected to benefit cancer patients. Recently, we engineered various peptide-Fc fusion proteins for directing Fcg receptor-bearing immune cells tow...

Descripción completa

Detalles Bibliográficos
Autores principales: Mobergslien, Anne, Peng, Qian, Vasovic, Vlada, Sioud, Mouldy
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5342789/
https://www.ncbi.nlm.nih.gov/pubmed/27713158
http://dx.doi.org/10.18632/oncotarget.12445
_version_ 1782513255420264448
author Mobergslien, Anne
Peng, Qian
Vasovic, Vlada
Sioud, Mouldy
author_facet Mobergslien, Anne
Peng, Qian
Vasovic, Vlada
Sioud, Mouldy
author_sort Mobergslien, Anne
collection PubMed
description Therapeutic strategies aiming at mobilizing immune effector cells to kill tumor cells independent of tumor mutational load and MHC expression status are expected to benefit cancer patients. Recently, we engineered various peptide-Fc fusion proteins for directing Fcg receptor-bearing immune cells toward tumor cells. Here, we investigated the immunostimulatory and anti-tumor effects of one of the engineered Fc fusion proteins (WN-Fc). In contrast to the Fc control, soluble WN-Fc-1 fusion protein activated innate immune cells (e.g. monocytes, macrophages, dendritic cells, NK cells), resulting in cytokine production and surface display of the lytic granule marker CD107a on NK cells. An engineered Fc-fusion variant carrying two peptide sequences (WN-Fc-2) also activated immune cells and bound to various cancer cell types with high affinity, including the murine 4T1 breast carcinoma cells. When injected into 4T1 tumor-bearing BALB/c mice, both peptide-Fc fusions accumulated in tumor tissues as compared to other organs such as the lungs. Moreover, treatment of 4T1 tumor-bearing BALB/c mice by means of two intravenous injections of the WN-Fc fusion proteins inhibited tumor growth with WN-Fc-2 being more effective than WN-Fc-1. Treatment resulted in tumor infiltration by T cells and NK cells. These new engineered WN-Fc fusion proteins may be a promising alternative to existing immunotherapies for cancer.
format Online
Article
Text
id pubmed-5342789
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-53427892017-03-28 Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth Mobergslien, Anne Peng, Qian Vasovic, Vlada Sioud, Mouldy Oncotarget Research Paper Therapeutic strategies aiming at mobilizing immune effector cells to kill tumor cells independent of tumor mutational load and MHC expression status are expected to benefit cancer patients. Recently, we engineered various peptide-Fc fusion proteins for directing Fcg receptor-bearing immune cells toward tumor cells. Here, we investigated the immunostimulatory and anti-tumor effects of one of the engineered Fc fusion proteins (WN-Fc). In contrast to the Fc control, soluble WN-Fc-1 fusion protein activated innate immune cells (e.g. monocytes, macrophages, dendritic cells, NK cells), resulting in cytokine production and surface display of the lytic granule marker CD107a on NK cells. An engineered Fc-fusion variant carrying two peptide sequences (WN-Fc-2) also activated immune cells and bound to various cancer cell types with high affinity, including the murine 4T1 breast carcinoma cells. When injected into 4T1 tumor-bearing BALB/c mice, both peptide-Fc fusions accumulated in tumor tissues as compared to other organs such as the lungs. Moreover, treatment of 4T1 tumor-bearing BALB/c mice by means of two intravenous injections of the WN-Fc fusion proteins inhibited tumor growth with WN-Fc-2 being more effective than WN-Fc-1. Treatment resulted in tumor infiltration by T cells and NK cells. These new engineered WN-Fc fusion proteins may be a promising alternative to existing immunotherapies for cancer. Impact Journals LLC 2016-10-04 /pmc/articles/PMC5342789/ /pubmed/27713158 http://dx.doi.org/10.18632/oncotarget.12445 Text en Copyright: © 2016 Mobergslien et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Mobergslien, Anne
Peng, Qian
Vasovic, Vlada
Sioud, Mouldy
Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth
title Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth
title_full Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth
title_fullStr Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth
title_full_unstemmed Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth
title_short Cancer cell-binding peptide fused Fc domain activates immune effector cells and blocks tumor growth
title_sort cancer cell-binding peptide fused fc domain activates immune effector cells and blocks tumor growth
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5342789/
https://www.ncbi.nlm.nih.gov/pubmed/27713158
http://dx.doi.org/10.18632/oncotarget.12445
work_keys_str_mv AT mobergslienanne cancercellbindingpeptidefusedfcdomainactivatesimmuneeffectorcellsandblockstumorgrowth
AT pengqian cancercellbindingpeptidefusedfcdomainactivatesimmuneeffectorcellsandblockstumorgrowth
AT vasovicvlada cancercellbindingpeptidefusedfcdomainactivatesimmuneeffectorcellsandblockstumorgrowth
AT sioudmouldy cancercellbindingpeptidefusedfcdomainactivatesimmuneeffectorcellsandblockstumorgrowth