Abstract

BackgroundNew treatment options for ovarian cancer are urgently required. Tumor-associated macrophages (TAMs) are an attractive target for therapy; repolarizing TAMs from M2 (pro-tumor) to M1 (anti-tumor) phenotypes represents an important therapeutic goal. We have previously shown that upregulated NF-kappaB (NF-κB) signaling in macrophages promotes M1 polarization, but effects in the context of ovarian cancer are unknown. Therefore, we aimed to investigate the therapeutic potential of increasing macrophage NF-κB activity in immunocompetent mouse models of ovarian cancer.MethodsWe have generated a transgenic mouse model, termed IKFM, which allows doxycycline-inducible overexpression of a constitutively active form of IKK2 (cIKK2) specifically within macrophages. The IKFM model was used to evaluate effects of increasing macrophage NF-κB activity in syngeneic murine TBR5 and ID8-Luc models of ovarian cancer in two temporal windows: 1) in established tumors, and 2) during tumor implantation and early tumor growth. Tumor weight, ascites volume, ascites supernatant and cells, and solid tumor were collected at sacrifice. Populations of macrophages and T cells within solid tumor and/or ascites were analyzed by immunofluorescent staining and qPCR, and soluble factors in ascitic fluid were analyzed by ELISA. Comparisons of control versus IKFM groups were performed by 2-tailed Mann-Whitney test, and a P-value < 0.05 was considered statistically significant.ResultsIncreased expression of the cIKK2 transgene in TAMs from IKFM mice was confirmed at the mRNA and protein levels. Tumors from IKFM mice, regardless of the timing of doxycycline (dox) administration, demonstrated greater necrosis and immune infiltration than control tumors. Analysis of IKFM ascites and tumors showed sustained shifts in macrophage populations away from the M2 and towards the anti-tumor M1 phenotype. There were also increased tumor-infiltrating CD3+/CD8+ T cells in IKFM mice, accompanied by higher levels of CXCL9, a T cell activating factor secreted by macrophages, in IKFM ascitic fluid.ConclusionsIn syngeneic ovarian cancer models, increased canonical NF-κB signaling in macrophages promoted anti-tumor TAM phenotypes and increased cytotoxic T cell infiltration, which was sufficient to limit tumor progression. This may present a novel translational approach for ovarian cancer treatment, with the potential to increase responses to T cell-directed therapy in future studies.

Highlights

  • New treatment options for ovarian cancer are urgently required

  • There were increased tumor-infiltrating CD3+/CD8+ T cells in IKFM mice, accompanied by higher levels of C-X-C Motif Chemokine Ligand 9 (CXCL9), a T cell activating factor secreted by macrophages, in IKFM ascitic fluid

  • In syngeneic ovarian cancer models, increased canonical NF-κB signaling in macrophages promoted anti-tumor TumorAssociated Macrophage (TAM) phenotypes and increased cytotoxic T cell infiltration, which was sufficient to limit tumor progression

Read more

Summary

Introduction

New treatment options for ovarian cancer are urgently required. New therapeutic approaches are urgently required to treat these patients and support tumor surveillance. The therapeutic potential of targeting cells in the tumor microenvironment (TME) that support tumor growth and metastasis is being increasingly recognized. Overcoming T cell exhaustion by tumor cells via checkpoint inhibitor therapy has led to durable responses in some solid tumor types, but ovarian cancer patients have shown relatively poor response rates at least in part due to low numbers of tumor-infiltrating T cells [3]. The ability to exploit these large macrophage populations in patients who have poor responses to traditional treatments and adaptive immunotherapies could provide a new direction in cancer therapeutics [8,9,10,11]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call