Pharmacologic ACE-Inhibition Mitigates Radiation-Induced Pneumonitis by Suppressing ACE-Expressing Lung Myeloid Cells

Int J Radiat Oncol Biol Phys. 2022 May 1;113(1):177-191. doi: 10.1016/j.ijrobp.2022.01.023. Epub 2022 Jan 29.

Abstract

Purpose: Radiation-induced lung injury is a major dose-limiting toxicity for thoracic radiation therapy patients. In experimental models, treatment with angiotensin converting enzyme (ACE) inhibitors mitigates radiation pneumonitis; however, the mechanism of action is not well understood. Here, we evaluate the direct role of ACE inhibition on lung immune cells.

Methods and materials: ACE expression and activity were determined in the lung immune cell compartment of irradiated adult rats after either high dose fractionated radiation therapy to the right lung (5 fractions × 9 Gy) or a single dose of 13.5 Gy partial body irradiation. Mitigation of radiation-induced pneumonitis with the ACE-inhibitor lisinopril was evaluated in the 13.5 Gy rat partial body irradiation model. During pneumonitis, we characterized inflammation and immune cell content in the lungs and bronchoalveolar lavage fluid. In vitro mechanistic studies were performed using primary human monocytes and the human monocytic THP-1 cell line.

Results: In both the partial body irradiation and fractionated radiation therapy models, radiation increased ACE activity in lung immune cells. Treatment with lisinopril improved survival during radiation pneumonitis (P = .0004). Lisinopril abrogated radiation-induced increases in bronchoalveolar lavage fluid monocyte chemoattractant protein 1 (chemokine ligand 2) and MIP-1a cytokine levels (P < .0001). Treatment with lisinopril reduced both ACE expression (P = .006) and frequency of CD45+ CD11b+ lung myeloid cells (P = .004). In vitro, radiation injury acutely increased ACE activity (P = .045) and reactive oxygen species (ROS) generation (P = .004) in human monocytes, whereas treatment with lisinopril blocked radiation-induced increases in both ACE and ROS. Radiation-induced ROS generation was blocked by pharmacologic inhibition of either NADPH oxidase 2 (P = .012) or the type 1 angiotensin receptor (P = .013).

Conclusions: These data demonstrate radiation-induced ACE activation within the immune compartment promotes the pathogenesis of radiation pneumonitis, while ACE inhibition suppresses activation of proinflammatory immune cell subsets. Mechanistically, our in vitro data demonstrate radiation directly activates the ACE/type 1 angiotensin receptor pathway in immune cells and promotes generation of ROS via NADPH oxidase 2.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Angiotensin-Converting Enzyme Inhibitors / pharmacology
  • Angiotensin-Converting Enzyme Inhibitors / therapeutic use
  • Animals
  • Humans
  • Lisinopril / pharmacology
  • Lisinopril / therapeutic use
  • Lung / radiation effects
  • Monocytes
  • NADPH Oxidase 2 / metabolism
  • Peptidyl-Dipeptidase A / metabolism
  • Peptidyl-Dipeptidase A / therapeutic use
  • Radiation Injuries* / pathology
  • Radiation Pneumonitis* / drug therapy
  • Radiation Pneumonitis* / etiology
  • Radiation Pneumonitis* / prevention & control
  • Rats
  • Reactive Oxygen Species / metabolism
  • Receptors, Angiotensin / metabolism
  • Receptors, Angiotensin / therapeutic use

Substances

  • Angiotensin-Converting Enzyme Inhibitors
  • Reactive Oxygen Species
  • Receptors, Angiotensin
  • Lisinopril
  • NADPH Oxidase 2
  • Peptidyl-Dipeptidase A