Your browser doesn't support javascript.
loading
Increased canonical NF-kappaB signaling specifically in macrophages is sufficient to limit tumor progression in syngeneic murine models of ovarian cancer.
Hoover, Alyssa A; Hufnagel, Demetra H; Harris, Whitney; Bullock, Kennady; Glass, Evan B; Liu, Esther; Barham, Whitney; Crispens, Marta A; Khabele, Dineo; Giorgio, Todd D; Wilson, Andrew J; Yull, Fiona E.
Affiliation
  • Hoover AA; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
  • Hufnagel DH; Vanderbilt University School of Medicine, Nashville, TN, USA.
  • Harris W; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
  • Bullock K; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
  • Glass EB; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
  • Liu E; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
  • Barham W; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
  • Crispens MA; Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
  • Khabele D; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
  • Giorgio TD; Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University School of Medicine, St. Louis, MO, USA.
  • Wilson AJ; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
  • Yull FE; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
BMC Cancer ; 20(1): 970, 2020 10 07.
Article in En | MEDLINE | ID: mdl-33028251
ABSTRACT

BACKGROUND:

New 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.

METHODS:

We 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.

RESULTS:

Increased 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.

CONCLUSIONS:

In 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.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: NF-kappa B / Macrophages Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Journal: BMC Cancer Journal subject: NEOPLASIAS Year: 2020 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: NF-kappa B / Macrophages Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Journal: BMC Cancer Journal subject: NEOPLASIAS Year: 2020 Document type: Article Affiliation country: United States