4.8 Article

Blockade of beta-adrenergic receptors reduces cancer growth and enhances the response to anti-CTLA4 therapy by modulating the tumor microenvironment

Journal

ONCOGENE
Volume 41, Issue 9, Pages 1364-1375

Publisher

SPRINGERNATURE
DOI: 10.1038/s41388-021-02170-0

Keywords

-

Funding

  1. Lundbeck Foundation [R307-2018-3326]
  2. Danish Cancer Society [R231-A14035, R231-A13832]
  3. Independent Research Fund Denmark
  4. department of Oncology, Herlev Hospital

Ask authors/readers for more resources

The development of immune checkpoint inhibitors (ICI) has been an important breakthrough in cancer therapies, but only a limited number of patients benefit from them. Recently, the nonselective beta blocker propranolol has been shown to be effective in treating angiosarcoma and soft tissue sarcomas. It works by reducing tumor angiogenesis and promoting an anti-tumoral microenvironment with increased T cell infiltration. The combination of propranolol with immune checkpoint inhibitors can significantly enhance their efficacy.
The development of immune checkpoint inhibitors (ICI) marks an important breakthrough of cancer therapies in the past years. However, only a limited fraction of patients benefit from such treatments, prompting the search for immune modulating agents that can improve the therapeutic efficacy. The nonselective beta blocker, propranolol, which for decades has been prescribed for the treatment of cardiovascular conditions, has recently been used successfully to treat metastatic angiosarcoma. These results have led to an orphan drug designation by the European Medicines Agency for the treatment of soft tissue sarcomas. The anti-tumor effects of propranolol are suggested to involve the reduction of cancer cell proliferation as well as angiogenesis. Here, we show that oral administration of propranolol delays tumor progression of MCA205 fibrosarcoma model and MC38 colon cancer model and increases the survival rate of tumor bearing mice. Propranolol works by reducing tumor angiogenesis and facilitating an anti-tumoral microenvironment with increased T cell infiltration and reduced infiltration of myeloid-derived suppressor cells (MDSCs). Using T cell deficient mice, we demonstrate that the full anti-tumor effect of propranolol requires the presence of T cells. Flow cytometry-based analysis and RNA sequencing of FACS-sorted cells show that propranolol treatment leads to an upregulation of PD-L1 on tumor associated macrophages (TAMs) and changes in their chemokine expression profile. Lastly, we observe that the co-administration of propranolol significantly enhances the efficacy of anti-CTLA4 therapy. Our results identify propranolol as an immune modulating agent, which can improve immune checkpoint inhibitor therapies in soft tissue sarcoma patients and potentially in other cancers.

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