4.8 Article

Macrophage-Derived Cholesterol Contributes to Therapeutic Resistance in Prostate Cancer

Journal

CANCER RESEARCH
Volume 81, Issue 21, Pages 5477-5490

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/0008-5472.CAN-20-4028

Keywords

-

Categories

Funding

  1. George Edgecomb Society and Department of Defense award [W81XWH-19-1-0435-PC 18103]
  2. NIH/NCI [U54CA193489, R00CA185325, R01CA230610]
  3. V Foundation
  4. 2021 Miles for Moffitt Award
  5. Moffitt Cancer Center
  6. Moffitt Cancer Center Proteomics and Metabolomics Core Facility by the NCI [P30-CA076292]
  7. Small Animal Imaging Lab Core Facility by the NCI [P30-CA076292]
  8. Flow Cytometry Core Facility by the NCI [P30-CA076292]
  9. Molecular Genomics Core Facility by the NCI [P30-CA076292]
  10. Analytic Microscopy Core Facility by the NCI [P30-CA076292]
  11. Tissue Core Facility by the NCI [P30-CA076292]

Ask authors/readers for more resources

The study shows that tumor-associated macrophages play a crucial role in CRPC by regulating cholesterol metabolism to impact androgen biosynthesis.
Castration-resistant prostate cancer (CRPC) is a lethal stage of disease in which androgen receptor (AR) signaling is persistent despite androgen deprivation therapy (ADT). Most studies have focused on investigating cell-autonomous alterations in CRPC, while the contributions of the tumor microenvironment are less well understood. Here we sought to determine the role of tumorassociated macrophages in CRPC, based upon their role in cancer progression and therapeutic resistance. In a syngeneic model that reflected the mutational landscape of CRPC, macrophage depletion resulted in a reduced transcriptional signature for steroid and bile acid synthesis, indicating potential perturbation of cholesterol metabolism. As cholesterol is the precursor of the five major types of steroid hormones, we hypothesized that macrophages were regulating androgen biosynthesis within the prostate tumor microenvironment. Macrophage depletion reduced androgen levels within prostate tumors and restricted AR nuclear localization in vitro and in vivo. Macrophages were also cholesterol-rich and were able to transfer cholesterol to tumor cells in vitro. AR nuclear translocation was inhibited by activation of liver X receptor (LXR)-b, the master regulator of cholesterol homeostasis. Consistent with these data, macrophage depletion extended survival during ADT and the presence of macrophages correlated with therapeutic resistance in patient-derived explants. Taken together, these findings support the therapeutic targeting of macrophages in CRPC. Significance: These results suggest that macrophage-targeted therapies can be combined with androgen deprivation therapy to treat patients with prostate cancer by limiting cholesterol bioavailability and the production of intratumoral androgens.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available