Monoclonal Antibody Requires Immunomodulation for Efficacy Against Acinetobacter baumannii Infection

TB Nielsen, J Yan, BM Luna, Y Talyansky… - The Journal of …, 2021 - academic.oup.com
The Journal of Infectious Diseases, 2021academic.oup.com
Monoclonal antibodies (mAbs) are gaining significant momentum as novel therapeutics for
infections caused by antibiotic-resistant bacteria. We evaluated the mechanism by which
antibacterial mAb therapy protects against Acinetobacter baumannii infections. Anticapsular
mAb enhanced macrophage opsonophagocytosis and rescued mice from lethal infections
by harnessing complement, macrophages, and neutrophils; however, the degree of bacterial
burden did not correlate with survival. Furthermore, mAb therapy reduced proinflammatory …
Abstract
Monoclonal antibodies (mAbs) are gaining significant momentum as novel therapeutics for infections caused by antibiotic-resistant bacteria. We evaluated the mechanism by which antibacterial mAb therapy protects against Acinetobacter baumannii infections. Anticapsular mAb enhanced macrophage opsonophagocytosis and rescued mice from lethal infections by harnessing complement, macrophages, and neutrophils; however, the degree of bacterial burden did not correlate with survival. Furthermore, mAb therapy reduced proinflammatory (interleukin-1β [IL-1β], IL-6, tumor necrosis factor-α [TNF-α]) and anti-inflammatory (IL-10) cytokines, which correlated inversely with survival. Although disrupting IL-10 abrogated the survival advantage conferred by the mAb, IL-10–knockout mice treated with mAb could still survive if TNF-α production was suppressed directly (via anti–TNF-α neutralizing antibody) or indirectly (via macrophage depletion). Thus, even for a mAb that enhances microbial clearance via opsonophagocytosis, clinical efficacy required modulation of pro- and anti-inflammatory cytokines. These findings may inform future mAb development targeting bacteria that trigger the sepsis cascade.
Oxford University Press