Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β

Background Metastatic breast cancer is a leading cause of cancer-related death in women worldwide. Infusion of natural killer (NK) cells is an emerging immunotherapy for such malignant tumors, although elimination of the immunosuppressive tumor environment is required to improve its efficacy. The ef...

Full description

Bibliographic Details
Main Authors: Demi Brownlie, Dahlia Doughty-Shenton, Daniel YH Soong, Colin Nixon, Neil O Carragher, Leo M Carlin, Takanori Kitamura
Format: Article
Language:English
Published: BMJ Publishing Group 2021-01-01
Series:Journal for ImmunoTherapy of Cancer
Online Access:https://jitc.bmj.com/content/9/1/e001740.full
id doaj-0a208b9ae45c43df991c0d62fdbd681b
record_format Article
spelling doaj-0a208b9ae45c43df991c0d62fdbd681b2021-07-14T09:30:24ZengBMJ Publishing GroupJournal for ImmunoTherapy of Cancer2051-14262021-01-019110.1136/jitc-2020-001740Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor βDemi Brownlie0Dahlia Doughty-Shenton1Daniel YH Soong2Colin Nixon3Neil O Carragher4Leo M Carlin5Takanori Kitamura6MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UKMRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UKMRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UKCancer Research UK Beatson Institute, Glasgow, UKCancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, The University of Edinburgh, Edinburgh, UKCancer Research UK Beatson Institute, Glasgow, UKMRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UKBackground Metastatic breast cancer is a leading cause of cancer-related death in women worldwide. Infusion of natural killer (NK) cells is an emerging immunotherapy for such malignant tumors, although elimination of the immunosuppressive tumor environment is required to improve its efficacy. The effects of this “metastatic” tumor environment on NK cells, however, remain largely unknown. Previous studies, including our own, have demonstrated that metastasis-associated macrophages (MAMs) are one of the most abundant immune cell types in the metastatic tumor niche in mouse models of metastatic breast cancer. We thus investigated the effects of MAMs on antitumor functions of NK cells in the metastatic tumor microenvironment.Methods MAMs were isolated from the tumor-bearing lung of C57BL/6 mice intravenously injected with E0771-LG mouse mammary tumor cells. The effects of MAMs on NK cell cytotoxicity towards E0771-LG cells were evaluated in vitro by real-time fluorescence microscopy. The effects of MAM depletion on NK cell activation, maturation, and accumulation in the metastatic lung were evaluated by flow cytometry (CD69, CD11b, CD27) and in situ hybridization (Ncr1) using colony-stimulating factor 1 (CSF-1) receptor conditional knockout (Csf1r-cKO) mice. Finally, metastatic tumor loads in the chest region of mice were determined by bioluminescence imaging in order to evaluate the effect of MAM depletion on therapeutic efficacy of endogenous and adoptively transferred NK cells in suppressing metastatic tumor growth.Results MAMs isolated from the metastatic lung suppressed NK cell-induced tumor cell apoptosis in vitro via membrane-bound transforming growth factor β (TGF-β) dependent mechanisms. In the tumor-challenged mice, depletion of MAMs increased the percentage of activated (CD69+) and mature (CD11b+CD27–) NK cells and the number of Ncr1+ NK cells as well as NK cell-mediated tumor rejection in the metastatic site. Moreover, MAM depletion or TGF-β receptor antagonist treatment significantly enhanced the therapeutic efficacy of NK cell infusion in suppressing early metastatic tumor outgrowth.Conclusion This study demonstrates that MAMs are a main negative regulator of NK cell function within the metastatic tumor niche, and MAM targeting is an attractive strategy to improve NK cell-based immunotherapy for metastatic breast cancer.https://jitc.bmj.com/content/9/1/e001740.full
collection DOAJ
language English
format Article
sources DOAJ
author Demi Brownlie
Dahlia Doughty-Shenton
Daniel YH Soong
Colin Nixon
Neil O Carragher
Leo M Carlin
Takanori Kitamura
spellingShingle Demi Brownlie
Dahlia Doughty-Shenton
Daniel YH Soong
Colin Nixon
Neil O Carragher
Leo M Carlin
Takanori Kitamura
Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
Journal for ImmunoTherapy of Cancer
author_facet Demi Brownlie
Dahlia Doughty-Shenton
Daniel YH Soong
Colin Nixon
Neil O Carragher
Leo M Carlin
Takanori Kitamura
author_sort Demi Brownlie
title Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
title_short Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
title_full Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
title_fullStr Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
title_full_unstemmed Metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
title_sort metastasis-associated macrophages constrain antitumor capability of natural killer cells in the metastatic site at least partially by membrane bound transforming growth factor β
publisher BMJ Publishing Group
series Journal for ImmunoTherapy of Cancer
issn 2051-1426
publishDate 2021-01-01
description Background Metastatic breast cancer is a leading cause of cancer-related death in women worldwide. Infusion of natural killer (NK) cells is an emerging immunotherapy for such malignant tumors, although elimination of the immunosuppressive tumor environment is required to improve its efficacy. The effects of this “metastatic” tumor environment on NK cells, however, remain largely unknown. Previous studies, including our own, have demonstrated that metastasis-associated macrophages (MAMs) are one of the most abundant immune cell types in the metastatic tumor niche in mouse models of metastatic breast cancer. We thus investigated the effects of MAMs on antitumor functions of NK cells in the metastatic tumor microenvironment.Methods MAMs were isolated from the tumor-bearing lung of C57BL/6 mice intravenously injected with E0771-LG mouse mammary tumor cells. The effects of MAMs on NK cell cytotoxicity towards E0771-LG cells were evaluated in vitro by real-time fluorescence microscopy. The effects of MAM depletion on NK cell activation, maturation, and accumulation in the metastatic lung were evaluated by flow cytometry (CD69, CD11b, CD27) and in situ hybridization (Ncr1) using colony-stimulating factor 1 (CSF-1) receptor conditional knockout (Csf1r-cKO) mice. Finally, metastatic tumor loads in the chest region of mice were determined by bioluminescence imaging in order to evaluate the effect of MAM depletion on therapeutic efficacy of endogenous and adoptively transferred NK cells in suppressing metastatic tumor growth.Results MAMs isolated from the metastatic lung suppressed NK cell-induced tumor cell apoptosis in vitro via membrane-bound transforming growth factor β (TGF-β) dependent mechanisms. In the tumor-challenged mice, depletion of MAMs increased the percentage of activated (CD69+) and mature (CD11b+CD27–) NK cells and the number of Ncr1+ NK cells as well as NK cell-mediated tumor rejection in the metastatic site. Moreover, MAM depletion or TGF-β receptor antagonist treatment significantly enhanced the therapeutic efficacy of NK cell infusion in suppressing early metastatic tumor outgrowth.Conclusion This study demonstrates that MAMs are a main negative regulator of NK cell function within the metastatic tumor niche, and MAM targeting is an attractive strategy to improve NK cell-based immunotherapy for metastatic breast cancer.
url https://jitc.bmj.com/content/9/1/e001740.full
work_keys_str_mv AT demibrownlie metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
AT dahliadoughtyshenton metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
AT danielyhsoong metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
AT colinnixon metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
AT neilocarragher metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
AT leomcarlin metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
AT takanorikitamura metastasisassociatedmacrophagesconstrainantitumorcapabilityofnaturalkillercellsinthemetastaticsiteatleastpartiallybymembraneboundtransforminggrowthfactorb
_version_ 1721303101173923840