Activating a collaborative innate-adaptive immune response to control breast and ovarian cancer metastasis

Sun, Lijuan, Kees, Tim, Almeida, Ana Santos, Liu, Bodu, He, Xue-Yan, Ng, David, Spector, David, McNeish, Iain A, Gimotty, Phyllis, Adams, Sylvia, Egeblad, Mikala (July 2020) Activating a collaborative innate-adaptive immune response to control breast and ovarian cancer metastasis. BioRxiv. (Unpublished)

[thumbnail of 2020.Sun.breast_ovarian_metastasis.pdf] PDF
2020.Sun.breast_ovarian_metastasis.pdf

Download (3MB)
DOI: 10.1101/2020.07.13.200477

Abstract

Many cancers recruit monocytes/macrophages and polarize them into tumor-associated macrophages (TAMs). TAMs promote tumor growth and metastasis and inhibit cytotoxic T cells. Yet, macrophages can also kill cancer cells after polarization by e.g., lipopolysaccharide (LPS, a bacteria-derived toll-like receptor 4 [TLR4] agonist) and interferon gamma (IFNγ). They do so via nitric oxide (NO), generated by inducible NO synthase (iNOS). Altering the polarization of macrophages could therefore be a strategy for controlling cancer. Here, we show that monophosphoryl lipid A (MPLA, a derivative of LPS) with IFNγ activated macrophages isolated from metastatic pleural effusions of breast cancer patients to kill the corresponding patients’ cancer cells in vitro. Importantly, intratumoral injection of MPLA with IFNγ not only controlled local tumor growth but also reduced metastasis in mouse models of luminal and triple negative breast cancers. Furthermore, intraperitoneal administration of MPLA with IFNγ reprogrammed peritoneal macrophages, suppressed metastasis, and enhanced the response to chemotherapy in the ID8-p53−/− ovarian carcinoma mouse model. The combined MPLA+IFNγ treatment reprogrammed the immunosuppressive microenvironment to be immunostimulatory by recruiting leukocytes, stimulating type I interferon signaling, decreasing tumor-associated (CD206+) macrophages, increasing tumoricidal (iNOS+) macrophages, and activating cytotoxic T cells through macrophage-secreted interleukin 12 (IL-12) and tumor necrosis factor α (TNFα). Both macrophages and T cells were critical for the anti-metastatic effects of MPLA+IFNγ. MPLA and IFNγ are already used individually in clinical practice, so our strategy to engage the anti-tumor immune response, which requires no knowledge of unique tumor antigens, may be ready for near-future clinical testing.

Item Type: Paper
Subjects: diseases & disorders > cancer > cancer types > breast cancer
organs, tissues, organelles, cell types and functions > cell types and functions > cell types > immune cell
organs, tissues, organelles, cell types and functions > cell types and functions > cell types > immune cell
organs, tissues, organelles, cell types and functions > cell types and functions > cell types > immune cell
diseases & disorders > cancer > metastasis
diseases & disorders > cancer > cancer types > ovarian cancer
CSHL Authors:
Communities: CSHL labs > Egeblad lab
CSHL labs > Spector lab
CSHL Cancer Center Program > Cellular Communication in Cancer Program
CSHL Cancer Center Program > Gene Regulation and Inheritance Program
SWORD Depositor: CSHL Elements
Depositing User: CSHL Elements
Date: 14 July 2020
Date Deposited: 13 Jul 2021 20:30
Last Modified: 13 Jul 2021 20:30
URI: https://repository.cshl.edu/id/eprint/40286

Actions (login required)

Administrator's edit/view item Administrator's edit/view item
CSHL HomeAbout CSHLResearchEducationNews & FeaturesCampus & Public EventsCareersGiving